WO2005016970A2 - Fully human antibodies directed against the human insulin-like growth factor-1 receptor - Google Patents

Fully human antibodies directed against the human insulin-like growth factor-1 receptor Download PDF

Info

Publication number
WO2005016970A2
WO2005016970A2 PCT/US2004/013852 US2004013852W WO2005016970A2 WO 2005016970 A2 WO2005016970 A2 WO 2005016970A2 US 2004013852 W US2004013852 W US 2004013852W WO 2005016970 A2 WO2005016970 A2 WO 2005016970A2
Authority
WO
WIPO (PCT)
Prior art keywords
igf
antibody
seq
tumor
antibodies
Prior art date
Application number
PCT/US2004/013852
Other languages
French (fr)
Other versions
WO2005016970A3 (en
Inventor
Dale L. Ludwig
Original Assignee
Imclone Systems Incorporated
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Imclone Systems Incorporated filed Critical Imclone Systems Incorporated
Priority to CA2524305A priority Critical patent/CA2524305C/en
Priority to EP04775941.0A priority patent/EP1622942B1/en
Priority to JP2006514278A priority patent/JP2007535895A/en
Priority to ES04775941.0T priority patent/ES2527871T3/en
Priority to US10/555,407 priority patent/US7638605B2/en
Publication of WO2005016970A2 publication Critical patent/WO2005016970A2/en
Publication of WO2005016970A3 publication Critical patent/WO2005016970A3/en
Priority to US12/640,671 priority patent/US7968093B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell

Definitions

  • IGF-IR insulin-like growth factor receptor
  • the IGF-IR is located on the cell surface of most cell types and serves as the signaling molecule for growth factors IGF-I and IGF-II (collectively termed henceforth IGFs). IGF-IR also binds insulin, albeit at three orders of magnitude lower affinity than it binds to IGFs. IGF-IR is a pre-formed hetero-tetramer containing two alpha and two beta chains covalently linked by disulfide bonds. The receptor subunits are synthesized as part of a single polypeptide chain of 180kd, which is then proteolytically processed into alpha (130kd) and beta (95kd) subunits. The entire alpha chain is extracellular and contains the site for ligand binding.
  • the beta chain possesses the transmembrane domain, the tyrosine kinase domain, and a C-terminal extension that is necessary for cell differentiation and transformation, but is dispensable for mitogen signaling and protection from apoptosis.
  • IGF-IR is highly similar to the insulin receptor (IR), particularly within the beta chain sequence (70% homology). Because of this homology, recent studies have demonstrated that these receptors can form hybrids containing one DR. dimer and one IGF-IR dimer (Pandini et al., Clin. Cane. Res. 5:1935-19 (1999)).
  • hybrids occur in both normal and transformed cells and the hybrid content is dependent upon the concentration of the two homodimer receptors (IR and IGF-IR) within the cell, hi one study of 39 breast cancer specimens, although both IR and IGF-IR were over-expressed in all tumor samples, hybrid receptor content consistently exceeded the levels of both homo-receptors by approximately 3-fold (Pandini et al, Clin. Cane. Res. 5:1935-44 (1999)). Although hybrid receptors are composed of IR and IGF-IR pairs, the hybrids bind selectively to IGFs, with affinity similar to that of IGF-IR, and only weakly bind insulin (Siddle and Soos, The IGF System. Humana Press, pp. 199-225. 1999).
  • IGF-IIR mannose-6-phosphate
  • M6P mannose-6-phosphate
  • IGF-IIR Loss of the IGF-IIR in tumor cells can enhance growth potential through release of its antagonistic effect on the binding of IGF-II with the IGF-IR (Byrd et al., J. Biol. Chem. 274:24408-16 (1999)).
  • Endocrine expression of IGF-I is regulated primarily by growth hormone and produced in the liver, but recent evidence suggests that many other tissue types are also capable of expressing IGF-I. This ligand is therefore subjected to endocrine and paracrine regulation, as well as autocrine in the case of many types of tumor cells (Yu, H. and Rohan, , J. Nail. Cancer Inst. 92:1472-89 (2000)).
  • IGFBPs IGF binding proteins
  • IGFBPs can either enhance or inhibit the action of IGFs, as determined by the molecular structures of the binding proteins as a result of post-translational modifications. Their primary roles are for transport of IGFs, protection of IGFs from proteolytic degradation, and regulation of the interaction of IGFs with IGF-IR. Only about 1% of serum IGF-I is present as free ligand, the remainder is associated with IGFBPs (Yu, H. and Rohan, J., J. Natl. Cancer Inst.
  • IGFs ligands
  • the IGF-IR Upon binding of ligand (IGFs), the IGF-IR undergoes autophosphorylation at conserved tyrosine residues within the catalytic domain of the beta chain. Subsequent phosphorylation of additional tyrosine residues within the beta chain provides docking sites for the recruitment of downstream molecules critical to the signaling cascade.
  • the principle pathways for transduction of the IGF signal are mitogen activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) (reviewed in Blakesley et al., In: The IGF System. Humana Press. 143-163 (1999)).
  • MAPK mitogen activated protein kinase
  • PI3K phosphatidylinositol 3-kinase
  • the MAPK pathway is primarily responsible for the mitogenic signal elicited following IGFs stimulation and PI3K is responsible for the IGF- dependent induction of anti-apoptotic or survival processes.
  • a key role of IGF-IR signaling is its anti-apoptotic or survival function.
  • Activated IGF-IR signals PI3K and downstream phosphorylation of Akt, or protein kinase B.
  • Akt can effectively block, through phosphorylation, molecules such as BAD, which are essential for the initiation of programmed cell death, and inhibit initiation of apoptosis (Datta et al., Cell 91 :231-41 (1997)).
  • Apoptosis is an important cellular mechanism that is critical to normal developmental processes (Oppenheim, Annu. Rev. Neurosci. 14:453-501 (1991)). It is a key mechanism for effecting the elimination of severely damaged cells and reducing the potential persistence of mutagenic lesions that may promote tumorigenesis. To this end, it has been demonstrated that activation of IGFs signaling can promote the formation of spontaneous tumors in a mouse transgenic model (DiGiovanni et al., Cancer Res. 60:1561-70 (2000)). Furthermore, IGF over-expression can rescue cells from chemotherapy induced cell death and may be an important factor in tumor cell drug resistance (Gooch et al., Breast Cancer Res. Treat. 56:1-10 (1999)).
  • IGF-IR and its ligands IGFs
  • IGF-I and IGF-II have been shown to be strong mitogens for a wide variety of cancer cell lines including prostate (Nickerson et al., Cancer Res.
  • IGF-IR knockout derived mouse embryo fibroblasts A critical role of the IGF-IR in cell proliferation and transformation was demonstrated in experiments of IGF-IR knockout derived mouse embryo fibroblasts. These primary cells grow at significantly reduced rates in culture medium containing 10% serum and fail to transform by a variety of oncogenes including SN40 Large T (Sell et al., Mol. Cell. Biol. 3604-12 (1994)). Recently it was demonstrated that resistance to the drug Herceptin in some forms of breast cancer may be due to activation of IGF-IR signaling in those cancers (Lu et al., J. Natl. Cancer Inst. 93:1852-57 (2001)). Over- expression or activation of IGF-IR may therefore not only be a major determinant in tumorigenicity, but also in tumor cell drug resistance.
  • Activation of the IGF system has also been implicated in several pathological conditions besides cancer, including acromegaly (Drange and Melmed. In: The IGF System. Humana Press. 699-720 (1999)), retinal neovascularization (Smith et al., Nature Med. 12:1390-95 (1999)), and psoriasis (Wraight et al., Nature Biotech. 18:521-26 (2000)).
  • an antisense oligonucleotide preparation targeting the IGF-IR was effective in significantly inhibiting the hyperproliferation of epidermal cells in human psoriatic skin grafts in a mouse model, suggesting that anti-IGF-IR therapies may be an effective treatment for this chronic disorder.
  • a variety of strategies have been developed to inhibit the IGF-IR signaling pathway in cells.
  • Antisense oligonucleotides have been effective in vitro and in experimental mouse models, as shown above for psoriasis.
  • inhibitory peptides targeting the IGF-IR have been generated that possess anti-proliferative activity in vitro and in vivo (Pietrzkowski et al., Cancer Res.
  • Antibodies are attractive therapeutics chiefly because of they 1) can possess high selectivity for a particular protein antigen, 2) are capable of exhibiting high affinity binding to the antigen, 3) possess long half- lives in vivo, and, since they are natural immune products, should 4) exhibit low in vivo toxicity (Park and Smolen. In: Advances in Protein Chemistry. Academic Press. pp:360-421 (2001)).
  • Antibodies derived from non-human sources, e.g.: mouse may, however, effect a directed immune response against the therapeutic antibody, following repeated application, thereby neutralizing the antibody's effectiveness.
  • Fully human antibodies offer the greatest potential for success as human therapeutics since they would likely be less immunogenic than murine or chimeric antibodies in humans, similar to naturally occurring immuno-responsive antibodies. To this end, there is a need to develop high affinity human anti-IGF-IR monoclonal antibodies for therapeutic use.
  • the invention provides human monoclonal antibodies and fragments thereof that bind specifically to the human IGF-I receptor.
  • the antibodies have at least one property selected from (i) inhibits binding of IGF-I or IGF-II to IGF-IR, (ii) neutralizes activation of IGF-IR by IGF-I or IGF-II, (iii) reduces IGF-IR surface receptor by at least about 80%; and (iv) binds to IGF-IR with a Kd of about 3 x 10 "10 or less.
  • an antibody of the invention reduces IGF-IR surface receptor by at least about 85%, and more preferably by at least about 90%.
  • the antibodies inhibit ligand-mediated receptor autophosphorylation and downstream cellular signaling through the MAPK and Akt pathways.
  • Antibodies of the invention used alone or in combination with an anti-neoplastic agent, are particularly useful for treating neoplastic diseases and hyperprohferative disorders.
  • the invention provides isolated polynucleotides encoding the antibodies or fragments thereof, expression vectors comprising the polynucleotide sequences, and host cells for expression.
  • the invention provides pharmaceutical compositions and diagnostic and therapeutic methods for treatment of tumors and hyperprohferative disease. The methods can further comprise administration of an anti-neoplastic agent or treatment.
  • Figure 1 depicts the nucleotide sequence of the 2F8 heavy chain variable domain.
  • Figure 2 depicts the amino acid sequence of the 2F8 heavy chain variable domain. CDRs are in bold and underlined.
  • Figure 3 depicts the nucleotide sequence of the complete 2F8 heavy chain (underline: secretory signal sequence; italics: IgGl constant region).
  • Figure 4 depicts the amino acid sequence of the complete 2F8 heavy chain (underline: secretory signal sequence; bold: CDRs; italics: IgGl constant region).
  • Figure 5 depicts the nucleotide sequence of the 2F8 light chain variable domain.
  • Figure 6 depicts the amino acid sequence of the 2F8 light chain variable domain. CDRs are in bold and underlined.
  • Figure 7 depicts the nucleotide sequence of the complete 2F8 light chain (underline: secretory signal sequence; italics: IgGl constant region).
  • Figure 8 depicts the amino acid sequence of the complete 2F8 light chain (underline: secretory signal sequence; bold: CDRs; italics: IgGl constant region).
  • Figure 9 depicts the nucleotide sequence of the A12 light chain variable domain.
  • Figure 10 depicts the amino acid sequence of the A12 light chain variable domain. CDRs are in bold and underlined. [0025] Fig.
  • FIG. 11 depicts the nucleotide sequence of the complete A12 light chain (underline: secretory signal sequence; italics: IgGl constant region).
  • Figure 12 depicts the amino acid sequence of the complete A12 light chain (underline: secretory signal sequence; bold: CDRs; italics: IgGl constant region).
  • Figure 13 depicts V H and N L CDR sequences of antibodies 2F8 and A12. Differences between the V CDRS are underlined.
  • Figure 14 depicts the homology between 2F8 and A12 light chain variable region amino acid sequences. Sequences differences are boxed and CDRs are highlighted.
  • Figure 15 shows results of an assay that measures the capacity of antibodies 2F8 and A12 to block binding of IGF-I to immobilized soluble IGF-IR. A12 antibodies having kappa or lambda light chain constant regions were tested.
  • Figure 16 shows results of an assay that measures the capacity of antibodies 2F8 and A12 (lambda light chain) to block binding of IGF-I to MCF7 cells.
  • Figure 17 shows results of an assay that measures the capacity of antibodies 2F8 and A12 to block insulin binding to ZR75-I cells.
  • Figure 18 shows the effect of antibody A12 on 3 H-thymidine incorporation in a mitogenesis assay.
  • Figure 18 A MCF7 breast cancer cells; Figure 18B: BxPC-3 pancreatic cancer cells; Figure 18C: HT-29 colon cancer cells.
  • Figure 19 shows inhibition of IGF-I mediated receptor phosphorylation.
  • Panel A Inhibition in MCF7 breast cancer cells by antibodies A12 ' and 2F8.
  • Panel B Inhibition in HT-29 colorectal cancer cells and BxPC-3 pancreatic cancer cells by antibody A12.
  • Figure 20 shows inhibition of IGF-I mediated phosphorylation of downstream effector molecules by antibodies A12 and 2F8.
  • Panel A inhibition of phosphorylation of MAPK
  • Panel B inhibition of phosphorylation of Akt.
  • Figure 21 shows binding of antibody A12 to human and murine IGF-IR positive and negative cell lines.
  • MCF7 human breast cancer cells; R-: mouse embryo fibroblasts; HEL: human leukemia cells; Lewis Lung: mouse lung carcinoma cells.
  • Figure 22 shows receptor internalization. Panel A shows internalization of labeled antibody A12 following binding to IGF-IR on MCF7 cells. Panel B shows depletion of cell-surface associated IGF-IR. Panel C shows degradation of total cellular IGF-IR after prolonged treatment with A12.
  • Figure 23 shows inhibition of HT-29 human colon carcinoma growth in nude mice by antibody 2F8 and CPT-11 (irinotecan) alone or in combination.
  • Figure 24 shows the effect of antibody A12 on HT-29 human colorectal tumor growth in nude mice.
  • Figure 25 shows the effect of antibody A12 on MCF7 human breast cancer growth in nude mice.
  • Figure 26 shows inhibition of BxPC-3 pancreatic cancer xenografts in nude mice by antibody A 12, gemcitabine, or CPT-11 (irinotecan) alone, or in combination.
  • Figure 27 shows inhibition of HT-29 colorectal cancer xenografts in nude mice by antibody A12, paclitaxel, or CPT-11 (irinotecan) alone, or in combination.
  • the present invention provides antibodies and fragments thereof, specific for insulin-like growth factor-I receptor (IGF-IR), as well as isolated or purified polynucleotide sequences encoding the antibodies.
  • IGF-IR insulin-like growth factor-I receptor
  • human antibodies are provided.
  • the antibodies can also be used in combination with other chemical and biological agents, including, but not limited to, anti-neoplastic agents and/or agents that are inhibitors of other receptors or receptor substrates mediating cell growth.
  • the invention further relates to anti-neoplastic agents that are inhibitors of topoisomerase function. The choice of such agents is advantageous for use in therapeutic methods in combination with antibodies that are specific for IGF-IR.
  • Naturally occurring antibodies typically have two identical heavy chains and two identical light chains, with each light chain covalently linked to a heavy chain by an interchain disulfide bond and multiple disulfide bonds further link the two heavy chains to one another.
  • Individual chains can fold into domains having similar sizes (110-125 amino acids) and structures, but different functions.
  • the light chain can comprise one variable domain (VL) and/or one constant domain (C L ).
  • the heavy chain can also comprise one variable domain (N ⁇ ) and/or, depending on the class or isotype of antibody, three or four constant domains (C H I, C H 2, C R 3 and C R 4).
  • the isotypes are IgA, IgD, IgE, IgG, and IgM, with IgA and IgG further subdivided into subclasses or subtypes (IgA 1-2 and [0044]
  • the variable domains show considerable amino acid sequence variability from one antibody to the next, particularly at the location of the antigen-binding site.
  • Three regions, called hypervariable or complementarity-determining regions (CDRs) are found in each of N and N H , which are supported by less variable regions called frameworks.
  • CDRs hypervariable or complementarity-determining regions
  • the portion of an antibody consisting of N and NH domains is designated Fv (Fragment variable) and constitutes the antigen-binding site.
  • Single chain Fv is an antibody fragment containing a V L domain and a V H domain on one polypeptide chain, wherein the ⁇ terminus of one domain and the C terminus of the other domain are joined by a flexible linker (see, e.g., U.S. Pat. No. 4,946,778 (Ladner et al.); WO 88/09344, (Huston et al.).
  • WO 92/01047 (McCafferty et al.) describes the display of scFv fragments on the surface of soluble recombinant genetic display packages, such as bacteriophage.
  • the peptide linkers used to produce the single chain antibodies can be flexible peptides selected to assure that the proper three-dimensional folding and association of the V L and V H domains occurs.
  • the linker is generally 10 to 50 amino acid residues.
  • the linker is 10 to 30 amino acid residues. More preferably the linker is 12 to 30 amino acid residues. Most preferably is a linker of 15 to 25 amino acid residues.
  • An non-limiting example of such a linker peptides is (Gly-Gly-Gly-Gly-Ser) 3 (SEQ ID NO:33).
  • Fab fragment, antigen binding refers to the fragments of the antibody consisting of V L -C L and V H -C H 1 domains.
  • Fab fragment, antigen binding
  • Such a fragment generated by digestion of a whole antibody with papain does not retain the antibody hinge region by which two heavy chains are normally linked.
  • the fragment is monovalent and simply referred to as Fab.
  • digestion with pepsin results in a fragment that retains the hinge region.
  • Such a fragment with intact interchain disulfide bonds linking two heavy chains is divalent and is referred to as F(ab') 2 -
  • a monovalent Fab' results when the disulfide bonds of an F(ab') 2 are reduced (and the heavy chains are separated.
  • Fc Frametic crystallization
  • the Fc is associated with Fc receptor binding, activation of complement-mediated cytdtoxicity and antibody-dependent cellular-cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular-cytotoxicity
  • the hinge region separates the Fab and Fc portions of the antibody, providing for mobility of Fabs relative to each other and relative to Fc, as well as including multiple disulfide bonds for covalent linkage of the two heavy chains.
  • Antibody formats have been developed which retain binding specificity, but have other characteristics that may be desirable, including for example, bispecificity, multivalence (more than two binding sites), compact size (e.g., binding domains alone).
  • Single chain antibodies lack some or all of the constant domains of the whole antibodies from which they are derived. Therefore, they can overcome some of the problems associated with the use of whole antibodies. For example, single-chain antibodies tend to be free of certain undesired interactions between heavy-chain constant regions and other biological molecules. Additionally, single-chain antibodies are considerably smaller than whole antibodies and can have greater permeability than whole antibodies, allowing single- chain antibodies to localize and bind to target antigen-binding sites more efficiently. Furthermore, the relatively small size of single-chain antibodies makes them less likely to provoke an unwanted immune response in a recipient than whole antibodies.
  • Multiple single chain antibodies each single chain having one V H and one V L domain covalently linked by a first peptide linker, can be covalently linked by at least one or more peptide linker to form a multivalent single chain antibodies, which can be monospecific or multispecific.
  • Each chain of a multivalent single chain antibody includes a variable light chain fragment and a variable heavy chain fragment, and is linked by a peptide linker to at least one other chain.
  • the peptide linker is composed of at least fifteen amino acid residues. The maximum number of amino acid residues is about one hundred.
  • Two single chain antibodies can be combined to form a diabody, also known as a bivalent dimer.
  • Diabodies have two chains and two binding sites, and can be monospecific or bispecific.
  • Each chain of the diabody includes a V H domain connected to a V L domain.
  • the domains are connected with linkers that are short enough to prevent pairing between domains on the same chain, thus driving the pairing between complementary domains on different chains to recreate the two antigen-binding sites.
  • Three single chain antibodies can be combined to form triabodies, also known as trivalent trimers. Triabodies are constructed with the amino acid terminus of a V L or V H domain directly fused to the carboxyl terminus of a V L or V H domain, i.e., without any linker sequence.
  • the triabody has three Fv heads with the polypeptides arranged in a cyclic, head- to-tail fashion. A possible conformation of the triabody is planar with the three binding sites located in a plane at an angle of 120 degrees from one another. Triabodies can be monospecific, bispecific or trispecific.
  • antibodies of the invention and fragments thereof include, but are not limited to, naturally occurring antibodies, bivalent fragments such as (Fab') 2 , monovalent fragments such as Fab, single chain antibodies, single chain Fv (scFv), single domain antibodies, multivalent single chain antibodies, diabodies, triabodies, and the like that bind specifically with antigens.
  • the antibodies of the present invention and particularly the variable domains thereof may be obtained by methods known in the art. These methods include, for example, the immunological method described by Kohler and Milstein, Nature, 256: 495-497 (1975) and Campbell, Monoclonal Antibody Technology, The Production and Characterization of Rodent and Human Hybridomas, Burdon et al., Eds., Laboratory Techniques in Biochemistry and Molecular Biology, Volume 13, Elsevier Science Publishers, Amsterdam (1985); as well as by the recombinant DNA methods such as described by Huse et al., Science, 246, 1275-81 (1989).
  • the antibodies can also be obtained from phage display libraries bearing combinations of V H and V L domains in the form of scFv or Fab.
  • V H and V L domains can be encoded by nucleotides that are synthetic, partially synthetic, or naturally derived.
  • phage display libraries bearing human antibody fragments can be preferred.
  • Other sources of human antibodies are transgenic mice engineered to express human immunoglobulin genes.
  • Antibody fragments can be produced by cleaving a whole antibody, or by expressing DNA that encodes the fragment. Fragments of antibodies may be prepared by methods described by Lamoyi et al., J. Immunol. Methods, 56: 235-243 (1983) and by Parham, J. Immunol. 131: 2895-2902 (1983). Such fragments may contain one or both Fab fragments or the F(ab') 2 fragment.
  • Such fragments may also contain single-chain fragment variable region antibodies, i.e. scFv, dibodies, or other antibody fragments.
  • Methods of producing such functional equivalents are disclosed in PCT Application WO 93/21319, European Patent Application No. 239,400; PCT Application WO 89/09622; European Patent Application 338,745; and European Patent Application EP 332,424.
  • the antibodies, or fragments thereof, of the present invention are specific for IGF-IR.
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen.
  • Antibodies, or fragments thereof, of the present invention for example, can be monospecific or bispecific.
  • Bispecific antibodies are antibodies that have two different antigen-binding specificities or sites. Where an antibody has more than one specificity, the recognized epitopes can be associated with a single antigen or with more than one antigen.
  • the present invention provides bispecific antibodies, or fragments thereof, that bind to two different antigens, with at least one specificity for IGF-IR.
  • Specificity of the present antibodies, or fragments thereof, for IGF-IR can be determined based on affinity and/or avidity. Affinity, represented by the equilibrium constant for the dissociation of an antigen with an antibody (Kd), measures the binding strength between an antigenic determinant and an antibody-binding site. Avidity is the measure of the strength of binding between an antibody with its antigen.
  • Avidity is related to both the affinity between an epitope with its antigen binding site on the antibody, and the valence of the antibody, which refers to the number of antigen binding sites specific for a particular epitope.
  • Antibodies typically bind with a dissociation constant (Kd) of IO "5 to 10 "11 liters/mol. Any Kd greater than IO "4 liters/mol is generally considered to indicate nonspecific binding. The lesser the value of the Kd, the stronger the binding strength between an antigenic determinant and the antibody binding site.
  • Kd dissociation constant
  • Antibodies of the present invention, or fragments thereof also include those for which binding characteristics have been improved by direct mutation, methods of affimty maturation, phage display, or chain shuffling.
  • Affinity and specificity can be modified or improved by mutating CDR and/or FW residues and screening for antigen binding sites having the desired characteristics (see, e.g., Yang et al., J. Mol. Biol., (1995) 254: 392-403).
  • One way is to randomize individual residues or combinations of residues so that in a population of, otherwise identical antigen binding sites, subsets of from two to twenty amino acids are found at particular positions.
  • mutations can be induced over a range of residues by error prone PCR methods (see, e.g., Hawkins et al., J. Mol. Biol, (1992) 226: 889-96).
  • phage display vectors containing heavy and light chain variable region genes can be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol., (1996) 250: 359-68). These methods of mutagenesis are illustrative of the many methods known to one of skill in the art.
  • Equivalents of the antibodies, or fragments thereof, of the present invention include polypeptides with amino acid sequences substantially the same as the amino acid sequence of the variable or hypervariable regions of the full-length anti-IGF-IR antibodies.
  • Substantially the same amino acid sequence is defined herein as a sequence with at least 70%, preferably at least about 80%, and more preferably at least about 90% homology to another amino acid sequence, as determined by the FASTA search method in accordance with Pearson and Lipman (Proc. Natl. Acad. Sci. USA (1988) 85: 2444-8).
  • Conservative amino acid substitution is defined as a change in the amino acid composition by way of changing one or two amino acids of a peptide, polypeptide or protein, or fragment thereof.
  • substitution is of amino acids with generally similar properties (e.g., acidic, basic, aromatic, size, positively or negatively charged, polarity, non-polarity) such that the substitutions do not substantially alter peptide, polypeptide or protein characteristics (e.g., charge, isoelectric point, affinity, avidity, conformation, solubility) or activity.
  • Typical substitutions that may be performed for such conservative amino acid substitution may be among the groups of amino acids as follows: glycine (G), alanine (A), valine (V), leucine (L) and isoleucine (I); aspartic acid (D) and glutamic acid (E); alanine (A), serine (S) and threonine (T); histidine (H), lysine (K) and arginine (R): asparagine (N) and glutamine (Q); phenylalanine (F), tyrosine (Y) and tryptophan (W) [0063] Conservative amino acid substitutions can be made in, e.g., regions flanking the hypervariable regions primarily responsible for the selective and/or specific binding characteristics of the molecule, as well as other parts of the molecule, e.g., variable heavy chain cassette.
  • Each domain of the antibodies of this invention can be a complete antibody with the heavy or light chain variable domain, or it can be a functional equivalent or a mutant or derivative of a naturally-occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Griffiths et al.). For instance, it is possible to join together domains corresponding to antibody variable domains, which are missing at least one amino acid.
  • the important characterizing feature is the ability of each domain to associate with a complementary domain to form an antigen- binding site. Accordingly, the terms variable heavy and light chain fragment should not be construed to exclude variants that do not have a material effect on specificity.
  • the anti-IGF-IR antibodies of the present invention are human antibodies that exhibit one or more of following properties.
  • the antibodies bind to the external domain of IGF-IR and inhibit binding of IGF-I or IGF-II to IGF-IR. Inhibition can be determined, for example, by a direct binding assay using purified or membrane bound receptor.
  • the antibodies of the present invention, or fragments thereof preferably bind IGF-IR at least as strongly as the natural ligands of IGF-IR (IGF-I and IGF-II).
  • the antibodies neutralize IGF-IR. Binding of a ligand, e.g.
  • IGF-I or IGF- II stimulates autophosphorylation of the beta subunit and phosphorylation of IFG-IR substrates, including MAPK, Akt, and IRS-1.
  • Neutralization of IGF-IR includes inhibition, diminution, inactivation and/or disruption of one or more of these activities normally associated with signal transduction. Further, this includes inhibition of IGF-IR / IR heterodimers as well as IGF-IR homodimers.
  • IGF-IR has various effects, including inhibition, diminution, inactivation and/or disruption of growth (proliferation and differentiation), angiogenesis (blood vessel recruitment, invasion, and metastasis), and cell motility and metastasis (cell adhesion and invasiveness).
  • One measure of IGF-IR neutralization is inhibition of the tyrosine kinase activity of the receptor. Tyrosine kinase inhibition can be determined using well-known methods; for example, by measuring the autophosphorylation level of recombinant kinase receptor, and/or phosphorylation of natural or synthetic substrates. Thus, phosphorylation assays are useful in determining neutralizing antibodies in the context of the present invention.
  • Phosphorylation can be detected, for example, using an antibody specific for phosphotyrosine in an ELISA assay or on a western blot.
  • Some assays for tyrosine kinase activity are described in Panek et al., J. Pharmacol. Exp. Thera. 283: 1433-44 (1997) and Batley et al., Life Sci. 62:143-50 (1998).
  • Antibodies of the invention cause a decrease in tyrosine phosphorylation of IGF-IR of at least about 75%, preferably at least about 85%, and more preferably at least about 90% in cells that respond to ligand.
  • IGF-IR neutralization Another measure of IGF-IR neutralization is inhibition of phosphorylation of downstream substrates of IGF-IR. Accordingly, the level of phosphorylation of MAPK, Akt, or IRS-1 can be measured. The decrease in substrate phosphorylation is at least about 50%, preferably at least about 65%, more preferably at least about 80%.
  • methods for detection of protein expression can be utilized to determine IGF-IR neutralization, wherein the proteins being measured are regulated by IGF- IR tyrosine kinase activity.
  • IHC immunohistochemistry
  • FISH fluorescence in situ hybridization
  • RT- PCR reverse transcriptase polymerase chain reaction
  • In vivo assays can also be utilized to determine IGF-IR neutralization.
  • receptor tyrosine kinase inhibition can be observed by mitogenic assays using cell lines stimulated with receptor ligand in the presence and absence of inhibitor.
  • MCF7 American Type Culture Collection (ATCC), Rockville, MD
  • IGF-I or IGF-II can be used to assay IGF-IR inhibition.
  • the present invention is not limited by any particular mechanism of IGF-IR neutralization.
  • the anti-IGF-IR antibodies of the present invention can bind externally to the IGF-IR cell surface receptor, block binding of ligand (e.g., IGF-I or IGF-II) and subsequent signal transduction mediated via the receptor-associated tyrosine kinase, and prevent phosphorylation of the IGF-IR and other downstream proteins in the signal transduction cascade.
  • ligand e.g., IGF-I or IGF-II
  • the antibodies down modulate IGF-IR.
  • the amount of IGF-IR present on the surface of a cell depends on receptor protein production, internalization, and degradation.
  • the amount of IGF-IR present on the surface of a cell can be measured indirectly, by detecting internalization of the receptor or a molecule bound to the receptor.
  • receptor internalization can be measured by contacting cells that express IGF-IR with a labeled antibody. Membrane-bound antibody is then stripped, collected and counted.
  • Internalized antibody is determined by lysing the cells and detecting label in the lysates.
  • Another way is to directly measure the amount of the receptor present on the cell following treatment with an anti-IGF-IR antibody or other substance, for example, by fluorescence-activated cell-sorting analysis of cells stained for surface expression of IGF-IR. Stained cells are incubated at 37°C and fluorescence intensity measured over time. As a control, part of the stained population can be incubated at 4°C (conditions under which receptor internalization is halted).
  • cell surface IGF-IR can be detected and measured using a different antibody that is specific for IGF-IR and that does not block or compete with binding of the antibody being tested.
  • IGF-IR expressing cell Treatment of an IGF-IR expressing cell with an antibody of the invention results in reduction of cell surface IGF-IR. In a preferred embodiment, the reduction is at least about 70%, more preferably at least about 80%, and even more preferably at least about 90% in response to treatment with an antibody of the invention. A significant decrease can be observed in as little as four hours.
  • Another measure of down-modulation is reduction of the total receptor protein present in a cell, and reflects degradation of internal receptors. Accordingly, treatment of cells (particularly cancer cells) with antibodies of the invention results in a reduction in total cellular IGF-IR.
  • the reduction is at least about 70%, more preferably at least about 80%, and even more preferably at least about 90%.
  • the antibodies of the invention bind to IGF-IR with a K d of about 3 x 10 "10 M "1 or less, preferably about 1 x 10 "10 M "1 or less, and more preferably about 3 x lO "11 E ar less.
  • the antibodies inhibit tumor growth.
  • subcutaneous xenograft tumors can be established by injection of cells of a cancer cell line into an immunodeficient mouse. The mice are then treated by intraperitoneal injection of antibodies, for example, every three days, and tumor size measured at regular intervals.
  • antibodies of the invention inhibit tumor growth.
  • an antibody of the invention promotes tumor regression when combined with an anti-neoplastic agent.
  • antibodies of the invention promoting tumor regression when used in a monotherapy.
  • promoting tumor regression is meant that administration of an effective amount of antibody, or an effective amount of a combination of an antibody and a neoplastic agent results in a reduction is size or necrosis of the tumor.
  • tumor regression may be observed and continue for a period of at least about 20 days, more preferably at least about 40 days, more preferably at least about 60 days.
  • Tumor regression may can be measured as an average across a group of subjects undergoing a particular treatment regimen, or can be measured by the number of subjects in a treatment group in which tumors regress.
  • Preferred antibodies of the present invention, or fragments thereof are human antibodies having one, two, three, four, five, and/or six complementarity determining regions (CDRs) selected from the group consisting of SEQ ID NO.14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28,and SEQ ID NO:30.
  • CDRs complementarity determining regions
  • the antibodies (or fragments thereof) of the present invention have CDRs of SEQ ID NO:14, SEQ ID NO:16 and SEQ ID NO:18.
  • the present antibodies, or fragments thereof have CDRs of SEQ ID NO:20, SEQ ID NO:22 and SEQ ID NO:24.
  • the present antibodies, or fragments thereof have CDRs of SEQ ID NO:26, SEQ ID NO:28 and SEQ ID NO:30.
  • the amino acid sequences of the CDRs are set forth below in Table 1.
  • the present antibodies, or fragments thereof can have a heavy chain variable region of SEQ ID NO:l and/or a light chain variable region selected from SEQ ID NO:5 or SEQ ID NO:6.
  • IMC-A12 is a particularly preferred antibody of the present invention. This antibody has human V H and V L framework regions (FWs) as well as CDRs.
  • the V H variable domain of IMC-A12 (SEQ ID NO:l) has three CDRs corresponding to SEQ ID NOs:14, 16, and 18 and the V L domain (SEQ ID NO:5) has three CDRs corresponding to SEQ ID NOS:20, 22, and 24.
  • IMC-2F8 is another preferred antibody of the present invention.
  • This antibody also has human V H and V L framework regions (FWs) and CDRs.
  • the V H variable domain of IMC-2F8 is identical to the V H variable domain of IMC- A12.
  • the V L domain of IMC-2F8 (SEQ ID NO:9) has three CDRs corresponding to SEQ ID NOS:26, 28, and 30.
  • antibodies of the invention compete for binding to IGF-IR with IMC-A12 and/or IMC-2F8. That is, the antibodies bind to the same or similar overlapping epitope.
  • the present invention also provides isolated polynucleotides encoding the antibodies, or fragments thereof, described previously.
  • the invention includes nucleic acids having a sequence encoding one, two, three, four, five and/or all six CDRs as set forth in Table 2.
  • TABLE 2 Heavy Chain (2F8/A12) CDR1 agctatgcta tcagc SEQ ID NO: 13 CDR2 gggatcatcc ctatctttgg tacagcaaac tacgcacaga SEQ ID NO: 15 agttccaggg c CDR3 gcgccattac gatttttgga gtggtccacc caagaccact SEQ ID NO: 17 actactacta ctacatg gacgtc Light Chain (2F8) CDR1 caaggagaca gcctcagaag ctattatgca age SEQ ID NO: 19 CDR2 ggtaaaaca accggccctc a SEQ ID N0:21 CDR3 aactcccggg acaaca
  • DNA encoding human antibodies can be prepared by recombining DNA encoding human constant regions and variable regions, other than the CDRs, derived substantially or exclusively from the corresponding human antibody regions and DNA encoding CDRs derived from a human (e.g., SEQ ID NOs:13, 15, and 17 for the heavy chain variable domain CDRs and SEQ ID NOs:19, 21, and 23 or SEQ ID NOS:25, 27 and 29 for the light chain variable domain CDRs).
  • Other suitable sources of DNAs that encode fragments of antibodies include any cell, such as hybridomas and spleen cells, that express the full-length antibody. The fragments may be used by themselves as antibody equivalents, or may be recombined into equivalents, as described above.
  • the present invention provides expression vectors containing the polynucleotide sequences previously described operably linked to an expression sequence, a promoter and an enhancer sequence.
  • expression vectors for the efficient synthesis of antibody polypeptide in prokaryotic, such as bacteria and eukaryotic systems, including but not limited to yeast and mammalian cell culture systems have been developed.
  • the vectors of the present invention can comprise segments of chromosomal, non- chromosomal and synthetic DNA sequences.
  • Any suitable expression vector can be used.
  • prokaryotic cloning vectors include plasmids from E. coli, such as colEl , pCRl , pBR322, pMB9, pUC, pKSM, and RP4.
  • Prokaryotic vectors also include derivatives of phage DNA such as MI 3 and other filamentous single-stranded DNA phages.
  • An example of a vector useful in yeast is the 2 ⁇ plasmid.
  • Suitable vectors for expression in mammalian cells include well-known derivatives of SV-40, adenovirus, retrovirus-derived DNA sequences and shuttle vectors derived from combination of functional mammalian vectors, such as those described above, and functional plasmids and phage DNA.
  • Additional eukaryotic expression vectors are known in the art (e.g., PJ. Southern and P. Berg, J. Mol. Appl. Genet. lj . 327-41 (1982); Subramani et al., Mol. Cell. Biol. 1: 854-64 (1981); Kaufrnann and Sharp, "Amplification And Expression of Sequences Cotransfected with a Modular Dihydrofolate Reductase Complementary DNA Gene," J. Mol. Biol. 159: 601-21 (1982); Kaufrnann and Sharp, Mol. Cell. Biol.
  • the expression vectors useful in the present invention contain at least one expression control sequence that is operatively linked to the DNA sequence or fragment to be expressed.
  • the control sequence is inserted in the vector in order to control and to regulate the expression of the cloned DNA sequence.
  • Examples of useful expression control sequences are the lac system, the trp system, the tac system, the trc system, major operator and promoter regions of phage lambda, the control region of fd coat protein, the glycolytic promoters of yeast, e.g., the promoter for 3-phosphoglycerate kinase, the promoters of yeast acid phosphatase, e.g., Pho5, the promoters of the yeast alpha-mating factors, and promoters derived from polyoma, adenovirus, retro virus, and simian virus, e.g., the early and late promoters or SV40, and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells and their viruses or combinations thereof.
  • a suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7. Stinchcomb et al. Nature, 282: 39 (1979); Kingsman et al., Gene, 7: 141 (1979).
  • the trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones, Genetics, 85: 12 (1977). The presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Leu2-deficient yeast strains are complemented by known plasmids bearing the Leu2 gene.
  • the present invention also provides recombinant host cells containing the expression vectors previously described. Antibodies of the present invention can be expressed in cell lines other than in hybridomas. Nucleic acids, which comprise a sequence encoding a polypeptide according to the invention, can be used for transformation of a suitable mammalian host cell. [0092] Cell lines of particular preference are selected based on high level of expression, constitutive expression of protein of interest and minimal contamination from host proteins.
  • Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines, such as but not limited to, COS-7 cells, Chinese Hamster Ovary (CHO) cells, Baby Hamster Kidney (BHK) cells and many others including cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells. Suitable additional eukaryotic cells include yeast and other fungi.
  • Useful prokaryotic hosts include, for example, E. coli, such as E. coli SG-936, E. coli HB 101 , E. coli W3110, E. coli XI 776, E. coli X2282, E. coli DHI, and E.
  • These present recombinant host cells can be used to produce an antibody, or fragment thereof, by culturing the cells under conditions permitting expression of the antibody or fragment thereof and purifying the antibody or fragment thereof from the host cell or medium surrounding the host cell. Targeting of the expressed antibody or fragment for secretion in the recombinant host cells can be facilitated by inserting a signal or secretory leader peptide-encoding sequence (see, Shokri et al., Appl Microbiol Biotechnol. 60:654-64 (2003) Nielsen et al, Erot. Eng.
  • secretory leader peptide elements can be derived from either prokaryotic or eukaryotic sequences. Accordingly suitably, secretory leader peptides are used, being amino acids joined to the N- terminal end of a polypeptide to direct movement of the polypeptide out of the host cell cytosol and secretion into the medium.
  • the transformed host cells are cultured by methods known in the art in a liquid medium containing assimilable sources of carbon (carbohydrates such as glucose or lactose), nitrogen (amino acids, peptides, proteins or their degradation products such as peptones, ammonium salts or the like), and inorganic salts (sulfates, phosphates and/or carbonates of sodium, potassium, magnesium and calcium).
  • the medium furthermore contains, for example, growth-promoting substances, such as trace elements, for example iron, zinc, manganese and the like.
  • Another embodiment for the preparation of antibodies in the present invention is the expression of the nucleic acid encoding the antibody according to the invention in a transgenic animal that has a substantial portion of the human antibody producing genome inserted and is rendered deficient in the production of endogenous antibodies.
  • Transgenic animals include but not limited to mice, goat, and rabbit.
  • One further embodiment of the invention include expression of the antibody-coding gene in, for example, the mammary gland of the animal for secretion of the polypeptide during lactation.
  • high affimty anti-IGF-IR antibodies according to the present invention can be isolated from a phage display library constructed from human heavy chain and light chain variable region genes.
  • variable domain of the invention can be obtained from a peripheral blood lymphocyte that contains a rearranged variable region gene.
  • variable domain portions such as CDR and FW regions, can be derived from different human sequences. Over 90% of recovered clones after three rounds of selection are specific to IGF-IR. The binding affinities for IGF-IR of the screened Fabs can be in the nM range, which is as high as many bivalent anti- IGF-IR monoclonal antibodies produced using hybridoma technology.
  • Antibodies, and fragments thereof, of the present invention can be obtained, for example, from naturally occurring antibodies, or Fab or scFv phage display libraries.
  • Single domain antibodies can be obtained by selecting a V H or a V L domain from a naturally occurring antibody or hybridoma, or selected from a library of V H domains or a library of V L domains. It is understood that amino acid residues that are primary determinants of binding of single domain antibodies can be within Kabat defined CDRs, but may include other residues as well, such as, for example, residues that would otherwise be buried in the V H -V L interface of a V H -V L heterodimer. [0098] Antibodies of the present invention also include those for which binding characteristics have been improved by direct mutation, methods of affimty maturation, phage display, or chain shuffling.
  • Affinity and specificity may be modified or improved by mutating CDRs and screening for antigen binding sites having the desired characteristics (see, e.g., Yang et al., J. Mol. Biol., 254: 392-403 (1995)).
  • CDRs are mutated in a variety of ways. One way is to randomize individual residues or combinations of residues so that in a population of otherwise identical antigen binding sites, all twenty amino acids are found at particular positions. Alternatively, mutations are induced over a range of CDR residues by error prone PCR methods (see, e.g., Hawkins et al., J. Mol. Biol., 226: 889-896 (1992)).
  • phage display vectors containing heavy and light chain variable region genes may be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol., 250: 359-368 (1996)). These methods of mutagenesis are illustrative of the many methods known to one of skill in the art.
  • the protein used to identify IGF-IR binding antibodies of the invention is preferably IGF-RI and, more preferably, is the extracellular domain of IGF-RI.
  • the IGF-RI extracellular domain can be free or conjugated to another molecule.
  • the antibodies of this invention can be fused to additional amino acid residues. Such amino acid residues can be a peptide tag, perhaps to facilitate isolation.
  • anti-IGF-IR antibodies or antibody fragments can be chemically or biosynthetically linked to anti-tumor agents or detectable signal-producing agents.
  • antibodies of the invention are efficiently internalized upon binding to cells bearing IGF-IR.
  • Anti-tumor agents linked to an antibody include any agents which destroy or damage a tumor to which the antibody has bound or in the environment of the cell to which the antibody has bound.
  • an anti-tumor agent is a toxic agent such as a chemotherapeutic agent or a radioisotope.
  • chemotherapeutic agents are known to those skilled in the art and include anthracyclines (e.g. daunomycin and doxorubicin), methotrexate, vindesine, neocarzinostatin, cis-platinum, chlorambucil, cytosine arabinoside, 5-fluorouridine, melphalan, ricin and calicheamicin.
  • the chemotherapeutic agents are conjugated to the antibody using conventional methods (See, e.g., Hermentin and Seiler, Behringlnst. Mitt. 82:197-215(1988)).
  • Detectable signal-producing agents are useful in vivo and in vitro for diagnostic purposes.
  • the signal producing agent produces a measurable signal which is detectable by external means, usually the measurement of electromagnetic radiation.
  • the signal producing agent is an enzyme or chromophore, or emits light by fluorescence, phosphorescence or chemiluminescence.
  • Chromophores include dyes which absorb light in the ultraviolet or visible region, and can be substrates or degradation products of enzyme catalyzed reactions.
  • the invention further contemplates anti-IGF-IR antibodies or antibody fragments of the invention to which target or reporter moieties are linked.
  • Target moieties are first members of binding pairs.
  • Anti-tumor agents for example, are conjugated to second members of such pairs and are thereby directed to the site where the antigen-binding protein is bound.
  • biotin is conjugated to an antigen-binding protein of the invention, and thereby provides a target for an anti-tumor agent or other moiety which is conjugated to avidin or streptavidin.
  • biotin or another such moiety is linked to an antigen-binding protein of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin.
  • Suitable radioisotopes for use as anti-tumor agents are also known to those skilled in the art. For example, I or At is used.
  • isotopes are attached to the antibody using conventional techniques (See, e.g., Pedley et al., Br. J. Cancer 68, 69- 73(1993)).
  • the anti-tumor agent which is attached to the antibody is an enzyme which activates a prodrug.
  • a prodrug is administered which remains in its inactive form until it reaches the tumor site where it is converted to its cytotoxin form once the antibody complex is administered.
  • the antibody-enzyme conjugate is administered to the patient and allowed to localize in the region of the tissue to be treated. The prodrug is then administered to the patient so that conversion to the cytotoxic drug occurs in the region of the tissue to be treated.
  • the anti-tumor agent conjugated to the antibody is a cytokine such as interleukin-2 (IL-2), interleukin-4 (E -4) or tumor necrosis factor alpha (TNF- ⁇ ).
  • IL-2 interleukin-2
  • E -4 interleukin-4
  • TNF- ⁇ tumor necrosis factor alpha
  • the antibody targets the cytokine to the tumor so that the cytokine mediates damage to or destruction of the tumor without affecting other tissues.
  • the cytokine is fused to the antibody at the DNA level using conventional recombinant DNA techniques.
  • a method of treating tumor growth in a mammal by administering to the mammal an effective amount of an antibody as previously described is also provided by the present invention.
  • the IGF-IR signaling pathway has been extensively demonstrated to be a causative factor in the development of many types of cancer.
  • IGF-I and IGF-II have been shown to be strong mitogens for a wide variety of cancer cell lines, including prostate, breast, colon, myeloma, ovary, pancreas and lung. Further, highly metastatic cancer cells have been shown to express higher levels of IGF-IR and IGF-II than tumor cells less prone to metastasize.
  • Suitable tumors to be treated according to the present invention preferably express IGF-IR. While not intended to be bound to any particular mechanism, the diseases and conditions which can be treated or prevented by the present methods include, for example, those in which pathogenic angiogenesis or tumor growth is stimulated through an IGF-IR paracrine and/or autocrine loop.
  • anti-IGF-IR antibodies can be ' administered in combination with one or more other anti-neoplastic agents.
  • combination therapies see, e.g., U.S. Patent No. 6,217,866 (Schlessinger et al.) (Anti-EGFR antibodies in combination with anti-neoplastic agents); WO 99/60023 (Waksal et al.) (Anti- EGFR antibodies in combination with radiation).
  • Any suitable anti-neoplastic agent can be used, such as a chemotherapeutic agent, radiation or combinations thereof.
  • the anti- neoplastic agent can be an alkylating agent or an anti-metabolite.
  • alkylating agents include, but are not limited to, cisplatin, cyclophosphamide, melphalan, and dacarbazine.
  • anti-metabolites include, but not limited to, doxorubicin, daunorubicin, and paclitaxel, gemcitabine, and topoisomerase inhibitors irinotecan (CPT-11), aminocamptothecin, camptothecin, DX-8951f, and topotecan (topoisomerase I) and etoposide (VP-16) and teniposide (VM-26) (topoisomerase II).
  • the source of the radiation can be either external (external beam radiation therapy - EBRT) or internal (brachytherapy - BT) to the patient being treated.
  • the dose of anti- neoplastic agent administered depends on numerous factors, including, for example, the type of agent, the type and severity tumor being treated and the route of administration of the agent. It should be emphasized, however, that the present invention is not limited to any particular dose.
  • the anti-neoplastic agents which are presently known in the art or being evaluated can be grouped into a variety of classes including, for example, mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, anti survival agents, biological response modifiers, anti-hormones, and anti-angiogenesis agents.
  • mitotic inhibitors alkylating agents
  • anti-metabolites intercalating antibiotics
  • growth factor inhibitors growth factor inhibitors
  • cell cycle inhibitors enzymes
  • topoisomerase inhibitors anti survival agents
  • biological response modifiers anti-hormones
  • anti-hormones anti-hormones
  • anti-angiogenesis agents anti-neoplastic agents which are particularly effective anti-neoplastic agents when used in combination with antibodies that bind to IGF-IR.
  • the inhibitors can be inhibitors of topoisomerase I or topoisomerase II.
  • Topoisomerase I inhibitors include irinotecan (CPT-11), aminocamptothecin, camptothecin, DX-8951f, topotecan.
  • Topoisomerase II inhibitors include etoposide (VP-16), and teniposide (NM-26). Other substances are currently being evaluated with respect to topoisomerase inliibitory activity and effectiveness as anti-neoplastic agents.
  • the topoisomerase inhibitor is irinotecan (CPT-11).
  • the antibodies used in combination are antibodies of the invention that bind to IGF-IR and have at least one of the following properties: (i) inhibit binding of IGF-I or IGF-II to IGF-IR; (ii) neutralize activation of IGF- IR by IGF-I or IGF-II; (iii) reduce IGF-IR surface receptor; and bind to IGF-IR with a Kd of about 1 x 10 "10 M "1 or less, hi a more preferred embodiment, the antibodies to be used in combination with a topoisomerase inhibitor have the characteristics of the human antibodies set forth above. [0110] Anti-IGF-IR antibodies of the invention can be admimstered with antibodies that neutralize other receptors involved in tumor growth or angiogenesis.
  • an anti-IGF-IR antibody is used in combination with a receptor antagonist that binds specifically to EGFR.
  • a receptor antagonist that binds specifically to EGFR.
  • antigen-binding proteins that bind to the extracellular domain of EGFR and block binding of one or more of its ligands and/or neutralize ligand-induced activation of EGFR.
  • An EGFR antagonist can be an antibody that binds to EGFR or a ligand of EGFR and inhibits binding of EGFR to its ligand.
  • Ligands for EGFR include, for example, EGF, TGF- ⁇ , amphiregulin, heparin-binding EGF (HB-EGF) and betacellulin.
  • EGF and TGF- ⁇ are thought to be the main endogenous ligands that result in EGFR-mediated stimulation, although TGF- ⁇ has been shown to be more potent in promoting angiogenesis.
  • the EGFR antagonist can bind externally to the extracellular portion of EGFR, which can or can not inhibit binding of the ligand, or internally to the tyrosine kinase domain.
  • Examples of EGFR antagonists that bind EGFR include, without limitation, biological molecules, such as antibodies (and ftmctional equivalents thereof) specific for EGFR, and small molecules, such as synthetic kinase inhibitors that act directly on the cytoplasmic domain of EGFR. [0111] Another example of such a receptor is VEGFR.
  • an anti-IGF-IR antibody is used in combination with a VEGFR antagonist, h one embodiment of the invention, an anti-IGF-IR antibody is used in combination with a receptor antagonist that binds specifically to VEGFR-1 /Fit- 1 receptor. In another embodiment, an anti-IGF-IR antibody is used in combination with a receptor antagonist that binds specifically to VEGFR-2/KDR receptor.
  • antigen-binding proteins that bind to the extracellular domain of VEGFR-1 or VEGFR-2 and block binding by their ligands (VEGFR-2 is stimulated most strongly byVEGF; VEGFR-1 is stimulated most strongly by P1GF, but also by VEGF) and/or neutralize ligand-induced induced activation.
  • IMC-1121 is a human antibody that binds to and neutralizes VEGFR-2 (WO 03/075840; Zhu).
  • MAb 6.12 is a scFv that binds to soluble and cell surface-expressed VEGFR-1. ScFv 6.12 comprises the V and VH domains of mouse monoclonal antibody MAb 6.12.
  • a hybridoma cell line producing MAb 6.12 has been deposited as ATCC number PTA-3344 under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure and the regulations thereunder (Budapest Treaty).
  • Other examples of growth factor receptors involved in tumorigenesis are the receptors for platelet-derived growth factor (PDGFR), nerve growth factor (NGFR), and fibroblast growth factor (FGFR).
  • the IGF-IR antibody can be administered in combination with one or more suitable adjuvants, such as, for example, cytokines (IL-10 and IL-13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides. See, e.g., Larrivee et al, supra. It should be appreciated, however, that administration of only an anti- IGF-IR antibody is sufficient to prevent, inhibit, or reduce the progression of the tumor in a therapeutically effective manner.
  • suitable adjuvants such as, for example, cytokines (IL-10 and IL-13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides. See, e.g., Larrivee et al, supra. It should be appreciated, however, that administration of only an anti- IGF-IR antibody is sufficient to prevent, inhibit, or reduce the progression of the tumor in a therapeutically effective manner.
  • the anti-IGF-IR antibody is administered before, during, or after commencing therapy with another agent, as well as any combination thereof, i.e., before and during, before and after, during and after, or before, during and after commencing the anti-neoplastic agent therapy.
  • the anti-IGF-IR antibody can be administered between 1 and 30 days, preferably 3 and 20 days, more preferably between 5 and 12 days before commencing radiation therapy.
  • chemotherapy is administered concurrently with or, more preferably, subsequent to antibody therapy.
  • any suitable method or route can be used to administer anti-IGF-IR antibodies of the invention, and optionally, to co-administer anti- neoplastic agents and/or antagonists of other receptors.
  • the anti-neoplastic agent regimens utilized according to the invention include any regimen believed to be optimally suitable for the treatment of the patient's neoplastic condition. Different malignancies can require use of specific anti-tumor antibodies and specific anti-neoplastic agents, which will be determined on a patient to patient basis.
  • Routes of administration include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration.
  • an anti-IGF-IR antibody of the invention can be administered as a conjugate, which binds specifically to the receptor and delivers a toxic, lethal payload following ligand-toxin internalization.
  • the antibody-drug/small molecule conjugate can be directly linked to each other or via a linker, peptide or non-peptide.
  • an anti-IGF-IR antibody of the invention can be chemically or biosynthetically linked to one or more anti-neoplastic or anti-angiogenic agents.
  • the invention further contemplates anti-IGF-IR antibodies to which target or reporter moieties are linked.
  • Target moieties are first members of binding pairs.
  • Anti- neoplastic agents for example, are conjugated to second members of such pairs and are thereby directed to the site where the anti-IGF-IR antibody is bound.
  • a common example of such a binding pair is avidin and biotin.
  • biotin is conjugated to an anti-IGF-IR antibody, and thereby provides a target for an anti-neoplastic agent or other moiety, which is conjugated to avidin or streptavidin.
  • biotin or another such moiety is linked to an anti-IGF-IR antibody of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin.
  • Suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins.
  • the compositions of the injection can, as is well known in the art, be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the mammal.
  • the present invention also includes kits for inhibiting tumor growth and/or angiogenesis comprising a therapeutically effective amount of a human anti-IGF-IR antibody.
  • kits can further contain any suitable antagonist of, for example, another growth factor receptor involved in tumorigenesis or angiogenesis (e.g., EGFR, VEGFR-1 /Fit- 1, VEGFR-2, PDGFR, NGFR, FGFR, etc, as described above).
  • the kits of the present invention can further comprise an anti-neoplastic agent. Examples of suitable anti- neoplastic agents in the context of the present invention have been described herein.
  • the kits of the present invention can further comprise an adjuvant; examples have also been described above. [0121]
  • kits which contain antibodies of the present invention.
  • the present receptor antibodies thus can be used in vivo and in vitro for investigative, diagnostic, prophylactic, or treatment methods, which are well known in the art.
  • investigative, diagnostic, prophylactic, or treatment methods which are well known in the art.
  • Phage were prepared by growing library stock to log phase culture, rescuing with M13K07 helper phage, and amplifying overnight at 30°C in 2YTAK culture medium at containing ampicillin and kanamycin selection.
  • the resulting phage preparation was precipitated in 4% PEG/0.5M NaCI and resuspended in 3% milk/PBS.
  • the immobilized receptors were then incubated with phage preparation for 1 hour at room temperature. Afterwards, the tubes were washed 10 times with PBST (PBS containing 0.1% Tween-20) followed by 10 times with PBS.
  • the bound phage were eluted at RT for 10 min with 1 ml of a freshly prepared solution of 100 mM triethylamine.
  • the eluted phage were incubated with 10 ml of mid-log phase TGI cells at 37°C for 30 min stationary and 30 min shaking.
  • the infected TGI cells were pelleted and plated onto several large 2YTAG plates and incubated overnight at 30°C. All colonies that grew on the plates were scraped into 3 to 5 ml of 2YTA medium, mixed with glycerol (final concentration: 10%), aliquoted and stored at -70°C.
  • 100 ⁇ l of the phage stock was added to 25 ml of 2YTAG medium and grown to mid-log phase.
  • the culture was rescued with M13K07 helper phage, amplified, precipitated, and used for selection following the procedure described above, but with reduced concentration (5 ⁇ g/ml) of IGF-IR immobilized onto tubes and increasing the numbers of washes following the binding process. A total of two rounds of selection were performed. [0125] Individual TGI clones were picked and grown at 37°C in 96 well plates and rescued with M13K07 helper phage as described above. The amplified phage preparation was blocked with 1/6 volume of 18% milk/PBS at RT for 1 h and added to Maxi-sorb 96-well microtiter plates (Nunc) coated with IGF-IR (1 ⁇ g/ml x 100 ⁇ l).
  • the Fab gene insert of individual clones was PCR amplified using primers: PUC19 reverse (5'- AGCGGATAACAATTTCACACAGG-3'; SEQ ID NO:31) and fdtet seq (5'- GTCGTCTTTCCAGACGTTAGT-3'; SEQ ID NO:32) which are specific for sequences flanking the unique Fab gene regions within the phage vector.
  • PUC19 reverse 5'- AGCGGATAACAATTTCACACAGG-3'; SEQ ID NO:31
  • fdtet seq 5'- GTCGTCTTTCCAGACGTTAGT-3'; SEQ ID NO:32
  • Each amplified product was digested with a frequent-cutting enzyme, BstN I, and analyzed on a 3% agarose gel. A total of 25 distinct patterns were identified. DNA sequences of representative clones from each digestion pattern were determined by dideoxynucleotide sequencing.
  • Plasmids from individual clones exhibiting positive binding to IGF-IR and unique DNA profile were used to transform a nonsuppressor E.coli host HB2151. Expression of the Fab fragments in HB2151 was induced by culturing the cells in 2YTA medium containing 1 mM isopropyl- 1-thio- ⁇ -D-galactopyranoside (IPTG, Sigma) at 30°C.
  • a periplasmic extract of the cells was prepared by resuspending the cell pellet in 25 mM Tris (pH 7.5) containing 20% (w/v) sucrose, 200 mM NaCI, 1 mM EDTA and 0.1 mM PMSF, followed by incubation at 4°C with gentle shaking for 1 h. After centrifugation at 15,000 rpm for 15 min, the soluble Fab protein was purified from the supernatant by affinity chromatography using Protein G column followed the manufacturer's protocol (Amersham Pharmacia Biotech).
  • Candidate binding Fab clones were screened for competitive blocking of radiolabeled human IGF-I ligand to immobilized IGF-IR (lOOng/well) coated onto 96 strip- well plates.
  • Fab preparations were diluted and incubated with IGF-IR plates for 0.5-1 hour at room temperature in PBS/0.1%BSA. Forty 40 pM of 125 I-IGF-I was then added and the plates incubated an additional 90 minutes. Wells were then washed 3 times with ice-cold PBS/0.1% BSA, dried, and then counted in a gamma scintillation counter.
  • the DNA sequence and translated amino acid sequence of the 2F8 light chain are shown in Figures 5 and 6, respectively.
  • the sequences for full-length lambda light chain format are shown in Figures 7 and 8.
  • Binding kinetic analysis was performed on 2F8 IgG using a BIAcore unit. This antibody was determined to bind to the IGF-IR with an affinity of 0.5 -1 nM (0.5-1 x IO "9 M).
  • a second generation Fab phage library was generated in which the 2F8 heavy chain was conserved and the light chain was varied to a diversity of greater than 10 unique species.
  • This method is termed light chain shuffling and has been used successfully to affinity mature selected antibodies for a given target antigen (Chames et al., J. Immunol. 169:1110-18 (2002)).
  • This library was then screened for binding to the human IGF-IR (10 ⁇ g/ml) following procedures as described above, and the panning process repeated an additional three rounds with reduced IGF-IR concentration (2 ⁇ g/ml) for enrichment of high affinity binding Fabs. Seven clones were analyzed following round four. All 7 contained the same DNA sequence and restriction digest profile.
  • the single isolated Fab was designated A12 and shown to possess a lambda light chain constant region.
  • the light chain DNA sequence is shown in Figure 9 and amino acid sequence in Figure 10.
  • Blocking results represent the level of antibody necessary to inhibit 50% binding of 1 * ⁇ I-IGF-I ligand to immobilized human IGF-IR. Affinity was determined by BIAcore analysis according to manufacturer's specifications (Pharmacia BIACORE 3000). Soluble IGF-IR was immobilized on the sensor chips and antibody binding kinetics determined.
  • the antibody changes incurred in 2F8 light chain to generate antibody A12 effected a significantly higher affinity of A12 for IGF-IR than 2F8. Concomitantly, this increase effected a greater binding ability of A12 for the receptor, as determined by ELISA, and at least a three-fold increase in blocking activity of ligand for immobilized receptor.
  • Figure 15 shows a representative titration of the two anti-IGF-IR antibodies in receptor blocking assay. The activity of A12 remained the same, irrespective of whether the light chain was engineered with a human lambda or kappa class constant region. Antibody A12 engineered with a lambda class light chain was utilized in all subsequent procedures.
  • the DNA sequences encoding the heavy and light chain genes of Fabs 2F8 and A12 were amplified by polymerase chain reaction (PCR) using the Boerhinger Mannheim Expand kit according to manufacturer's instructions. Forward and reverse primers contained sequences for restriction endonuclease sites for cloning into mammalian expression vectors.
  • the recipient vector for the heavy chain contained the entire human gamma 1 constant region cDNA sequence, flanked by a strong eukaryotic promoter and a 3 ' polyadenylation sequence.
  • the full-length lambda light chain sequences for 2F8 or A12 were each cloned in to a second vector possessing only the eukaryotic regulatory elements for expression in mammalian cells.
  • a selectable marker was also present on this vector for selection of stable DNA integrants following transfection of the plasmid into mammalian cells.
  • Forward primers were also engineered with sequences encoding a strong mammalian signal peptide sequence for proper secretion of the expressed antibody.
  • the DNAs were sequenced and tested for expression in transient transfection. Transient transfection was performed into the COS7 primate cell line using Lipofection, according to manufacturer's specifications.
  • ELISA Plates (96 well) were prepared by coating with 100 ng/well of a goat-anti-human Fc-specific polyclonal antibody (Sigma) and blocked with 5% milk/PBS overnight at 4°C. The plates were then washed 5 times with PBS. Conditioned supernatant was added tol wells and incubated for 1.5 hours at room temperature. Bound antibody was detected with a goat anti-human lambda light chain-HRP antibody (Sigma) and visualized with TMB reagents and microplate reader as described above.
  • Antibody generated by either transient or stable transfection was purified by ProA affinity chromatography (Harlow and Lane. Antibodies. A Laboratory Manual. Cold Spring Harbor Press. 1988), eluted into a neutral buffered saline solution, and quantitated.
  • I-IGF-I (approximately 40,000 cpm/well) was added to each well and incubated for an additional hour with gentle agitation. The wells were then washed three times with ice-cold PBS / 0.1% BSA. Monolayers were then lysed with 200 ⁇ l 0.5N NaOH and counted in a gamma counter. The results are shown graphically in Figure 16. On human tumor cells, antibody A12 inhibited ligand binding to IGF-IR with an IC 5 o of 3 nM (0.45 ⁇ g/ml).
  • the IGF-IR shares considerable homology with the insulin receptor (IR). To determine if the anti-IGF-IR antibodies were specific to this IGF-IR and did not block insulin binding, a cell-based blocking assay was performed on human ZR-75I breast cancer cells. Because insulin can bind to IGF-IR, albeit at three orders of magnitude lower affinity than for the IR, we utilized the human breast cancer line ZR-75I that possesses a higher IR to IGF-IR ratio in comparison to MCF7 cells. By using this line, we reasoned that insulin binding to the cells would be more indicative of specific IR binding. The assay was performed as described above for MCF7 cells and the results shown in Figure 17.
  • IGF-I insulin growth factor-I
  • Antibody A12 inhibited MCF7 mitogenesis in response to IGF-I ligand in a dose-dependent fashion, with an IC 50 of 6 nM.
  • Antibody A12 was then tested for mitogenic inhibition on several additional human tumor cells lines and the results shown in Table 5.
  • Antibody A12 was effective at inhibiting IGF-I ligand-mediated mitogenesis of a variety of human tumor cell lines, including breast cancer, colorectal cancer, and multiple myeloma.
  • Antibody-mediated inhibition of IGF-I directed receptor phosphorylation and downstream signalins were performed in the presence or absence of antibody A12 or 2F8.
  • the MCF7 human breast cancer cell line was selected for use due to its high IGF-IR density. Cells were plated into 10 cm or 6 well culture dishes and grown to 70-80% confluence. The monolayers were then washed twice in PBS and cultured overnight in serum free defined medium.
  • Anti- IGF-IR antibody was then added in fresh serum-free media (100 nM-10 nM) and incubated cells 30 minutes before addition of ligand (10 nM). Cells were incubated with ligand for 10 minutes, then placed on ice and washed with ice-cold PBS.
  • the cells were lysed by the addition of lysis solution (50 mM Tris-HCl, pH 7.4, 150 mM NaCI, 1% TritonX-100, 1 mM EDTA, 1 mM PMSF, 0.5 mM Na 3 VO 4 , 1 ⁇ g/ml leupeptin, 1 ⁇ g/ml pepstatin, and 1 ⁇ g/ml aprotinin) and the cells scraped into a centrifuge tube kept on ice for 15 minutes. The lysate was then clarified by centrifugation at 4°C. Solubilized IGF-IR was then immunoprecipitated (IP) from the lysate.
  • IP immunoprecipitated
  • Antibody 3B7 (Santa Cruz) or A12 at 1 ⁇ g/ml were incubated with 400 ⁇ l of lysate overnight at 4°C. Immune complexes were then precipitated by the addition of ProA-sepharose beads for 2 hours at 4°C, pelleted, and washed 3 times with lysis buffer. IP s bound to the ProA beads were stripped into denaturing gel running buffer. Lysate or IP were processed for denaturing gel electrophoresis and run on a 4-12% acrylamide gel and blotted to nylon or nitrocellulose membrane by western blot according to Towbin et al. (Biotechnology 24: 145-9 (1992)).
  • Tyrosine phosphorylated protein was detected on the blot using an anti-p-tyrosine antibody (Cell Signaling #9411) and an anti-mouse-HRP secondary antibody. IGF-IR was detected with monoclonal antibody C-20 (Santa Cruz). For Akt phosphorylation, phospho-Akt was detected with antibody #559029 and total Akt with #559028 (BD Pharmingen). For MAPK phosphorylation, phospho-p44/42 was detected with #9101 and total p44/42 with #9102 (Cell Signaling Tech.). Bands were visualized with the ECL reagent on X-ray film.
  • A12 was capable of immunoprecipitating endogenous IGF-IR, we were therefore interested in determining if A12 could also be used as detection antibody for fluorescence activated cell sorting (FACs).
  • FACs fluorescence activated cell sorting
  • A12 failed to bind to these cells, indicative of antibody binding specificity for the IGF-IR. A12 did, however, bind and partially shift the mouse tumor cell line Lewis Lung carcinoma, suggesting that this anti-human IGF-IR antibody possesses some cross-reactivity for the mouse IGF-IR.
  • Antibody A12 has been shown to bind native IGF-IR on human tumor cells with high affinity. Antibody A12 was radio-iodinated with 125 iodine using IODO-beads (Pierce) according to manufacturer's instructions. MCF7 human breast cancer cells were plated into 6-well plates and cultured overnight to 50% confluence. One microgram of 125 I- A12 was added to each well and incubated at 37°C or kept on ice at 4°C. Plates were incubated for 30 minutes, 90 minutes, or 180 minutes and each time point performed in triplicate. The culture at 4°C was held for 180 minutes.
  • IGF-IR surface receptor density was determined by FACS.
  • Adherent MCF 7 cells were treated for 4 h with 50 nM of antibody A12 of IGF-I at 37°C. Cells were washed in ice-cold PBS/5% BSA twice, and 1 x IO 6 cells were aliquoted to staining tubes and placed on ice.
  • An IGF-IR alpha-polypeptide specific, non-blocking mouse monoclonal antibody (Ab-1, NeoMarkers, Fremont, CA), was then incubated with cells at 4°C for 2 h.
  • the drug irenotecan (CPT-11) (LKT Laboratories) was injected IP (100 mg/kg) once a week for four weeks from the initiation of antibody treatment. Control animals received a class matched irrelevant human IgG antibody. Tumor measurements were performed at regular intervals using Vernier calipers, measuring height, width, and length and calculated to determine the total tumor volume. The study was terminated when control tumors reached 3000 mm . As shown in Fig. 23, doses of antibody 2F8 at either 0.5 mg or 1 mg every three days effected a significant inhibition (P ⁇ 0.05) of tumor growth in this model. There was no statistical difference between the tumor sizes from groups treated with 0.5 or 1 mg 2F8 and the responses were similar to treatment with CPT-11 alone. When 2F8 and CPT-11 were given together the combination resulted in greater inhibition of tumor growth (72% decrease), demonstrating that anti-IGF-IR therapy could enhance the anti-tumor activity of the chemotherapeutic agent CPT-11 on tumor growth.
  • Anti-tumor activity of antibody A12 on human colorectal tumors in vivo [0146] Antibody A12 possesses a 10-fold higher affinity for the IGF-IR than antibody 2F8. Since significant tumor inhibition was observed in vivo with antibody 2F8, we investigated the activity of A12 on the growth of the human colorectal cancer line HT-29 in a mouse xenograft model. Tumors were induced as previously described, and antibody treatment initiated once tumors were established (200 mm 3 size). Antibody treatment was then given at a concentration of 1 mg, 100 ⁇ g, or 10 ⁇ g every three days throughout the duration of the experiment. Ten animals were used per treatment group and control animals received a class match IgG control antibody. As shown in Fig.
  • antibody A12 effected a 74% reduction in tumor growth compared to control (P ⁇ 0.05). This demonstrated that A12 was effective as a single therapeutic at inhibiting colorectal tumor growth in this xenograft model. A clear dose-response effect was noted in this experiment. Anti-tumor activity was also observed with a dose of 100 ⁇ g A12.
  • Antibody A12 exhibited strong inhibitory activity on the IGF-dependent mitogenic stimulation and proliferation of MCF7 cells in vitro.
  • a mouse xenograft tumor model was utilized. MCF7 cells were originally isolated from an estrogen-dependent human tumor and require exogenously added estrogen for maintenance and growth in vivo. Nude mice were implanted with biodegradable estrogen pellets (0.72mg 17- ⁇ -estradiol/pellet, 60 day release).
  • mice were also injected in the right flank subcutaneously with 0.5 mg of estradiol in a 50 ⁇ l suspension of sesame seed oil. Tumors were allowed to establish a size of approximately 150 mm before antibody treatment was initiated. Antibody was injected at 1 mg, 100 ⁇ g, and 10 ⁇ g doses every three days and continued for the duration of the experiment. At 29 days, treatment of animals with 1 mg of A12 effected an 89% reduction in tumor growth (Fig. 25). Minimal growth was apparent for the established tumors in this treatment group. A dose dependent response was noted for A12 treatments in this model. The study demonstrated that A12 was effective in significantly reducing the growth of a human breast cancer cell line in vivo. Further, treating with antibody alone at 1 mg/dose, tumor regression was observed and continued to the termination of the study (50 days).
  • mice were injected subcutaneously with 2 x 10 s BxPC-3 human pancreatic carcinoma cells mixed 1 : 1 with Matrigel. Twenty days later, when tumors reached 200-300mm 3 , mice were randomized and divided into treatment groups: 1) TBS control; 2) mAb A12 at lmg/dose, 3 times per week; 3) 1 mg irinotecan, once every 7 days; 4) mAb A12 + irinotecan; 5) 2.5 mg gemcitabine, once every 7 days; 6) mAb A12 + gemcitabine.
  • Efficacy ofA12 in combination with CPT-11 or paclitaxel in HT-29 colorectal cancer xenografts [0151] Female thymic nu/nu mice were injected subcutaneously with HT-29 human colon carcinoma cell suspension, at 5 x 10 cells in 0.4 ml mixed 1:1 with Matrigel.

Abstract

This invention relates to human antibodies that bind to human insulin-like growth factor-1 receptor (IGF-IR), to derivatives of these antibodies (Fabs, single chain antibodies, bi-specific antibodes, or fusion proteins), and to uses of the antibodies and derivatives in therapeutic, and diagnostic methods. The invention relates to nucleic acids encoding the anti-IGF-IR, methods of generating the antibodies and expression. The invention further relates to combination therapies using ant-IGF-IR antibodies with anti-neoplastic drugs.

Description

FULLY HUMAN ANTIBODIES DIRECTED AGAINST THE HUMAN INSULIN-LIKE GROWTH FACTOR-1 RECEPTOR
[0001] This application claims the benefit of United States Provisional Application 60/467,177, filed May 1, 2003.
BACKGROUND [0002] The insulin-like growth factor receptor (IGF-IR) is a ubiquitous transmembrane tyrosine kinase receptor that is essential for normal fetal and post-natal growth and development. IGF-IR can stimulate cell proliferation, cell differentiation, changes in cell size, and protect cells from apoptosis. It has also been considered to be quasi- obligatory for cell transformation (reviewed in Adams et al., Cell. Mol. Life Sci. 57:1050-93 (2000); Baserga, Oncogene 19:5574-81 (2000)). The IGF-IR is located on the cell surface of most cell types and serves as the signaling molecule for growth factors IGF-I and IGF-II (collectively termed henceforth IGFs). IGF-IR also binds insulin, albeit at three orders of magnitude lower affinity than it binds to IGFs. IGF-IR is a pre-formed hetero-tetramer containing two alpha and two beta chains covalently linked by disulfide bonds. The receptor subunits are synthesized as part of a single polypeptide chain of 180kd, which is then proteolytically processed into alpha (130kd) and beta (95kd) subunits. The entire alpha chain is extracellular and contains the site for ligand binding. The beta chain possesses the transmembrane domain, the tyrosine kinase domain, and a C-terminal extension that is necessary for cell differentiation and transformation, but is dispensable for mitogen signaling and protection from apoptosis. [0003] IGF-IR is highly similar to the insulin receptor (IR), particularly within the beta chain sequence (70% homology). Because of this homology, recent studies have demonstrated that these receptors can form hybrids containing one DR. dimer and one IGF-IR dimer (Pandini et al., Clin. Cane. Res. 5:1935-19 (1999)). The formation of hybrids occurs in both normal and transformed cells and the hybrid content is dependent upon the concentration of the two homodimer receptors (IR and IGF-IR) within the cell, hi one study of 39 breast cancer specimens, although both IR and IGF-IR were over-expressed in all tumor samples, hybrid receptor content consistently exceeded the levels of both homo-receptors by approximately 3-fold (Pandini et al, Clin. Cane. Res. 5:1935-44 (1999)). Although hybrid receptors are composed of IR and IGF-IR pairs, the hybrids bind selectively to IGFs, with affinity similar to that of IGF-IR, and only weakly bind insulin (Siddle and Soos, The IGF System. Humana Press, pp. 199-225. 1999). These hybrids therefore can bind IGFs and transduce signals in both normal and transformed cells. [0004] A second IGF receptor, IGF-IIR, or mannose-6-phosphate (M6P) receptor, also binds IGF-II ligand with high affinity, but lacks tyrosine kinase activity (Oates et al., Breast Cancer Res. Treat. 47:269-81 (1998)). Because it results in the degradation of IGF-II, it is considered a sink for IGF-II, antagonizing the growth promoting effects of this ligand. Loss of the IGF-IIR in tumor cells can enhance growth potential through release of its antagonistic effect on the binding of IGF-II with the IGF-IR (Byrd et al., J. Biol. Chem. 274:24408-16 (1999)). [0005] Endocrine expression of IGF-I is regulated primarily by growth hormone and produced in the liver, but recent evidence suggests that many other tissue types are also capable of expressing IGF-I. This ligand is therefore subjected to endocrine and paracrine regulation, as well as autocrine in the case of many types of tumor cells (Yu, H. and Rohan, , J. Nail. Cancer Inst. 92:1472-89 (2000)). [0006] Six IGF binding proteins (IGFBPs) with specific binding affinities for the IGFs have been identified in serum (Yu, H. and Rohan, j., J. Natl. Cancer Inst. 92:1472-89 (2000)). IGFBPs can either enhance or inhibit the action of IGFs, as determined by the molecular structures of the binding proteins as a result of post-translational modifications. Their primary roles are for transport of IGFs, protection of IGFs from proteolytic degradation, and regulation of the interaction of IGFs with IGF-IR. Only about 1% of serum IGF-I is present as free ligand, the remainder is associated with IGFBPs (Yu, H. and Rohan, J., J. Natl. Cancer Inst. 92:1472-89 (2000)). [0007] Upon binding of ligand (IGFs), the IGF-IR undergoes autophosphorylation at conserved tyrosine residues within the catalytic domain of the beta chain. Subsequent phosphorylation of additional tyrosine residues within the beta chain provides docking sites for the recruitment of downstream molecules critical to the signaling cascade. The principle pathways for transduction of the IGF signal are mitogen activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) (reviewed in Blakesley et al., In: The IGF System. Humana Press. 143-163 (1999)). The MAPK pathway is primarily responsible for the mitogenic signal elicited following IGFs stimulation and PI3K is responsible for the IGF- dependent induction of anti-apoptotic or survival processes. [0008] A key role of IGF-IR signaling is its anti-apoptotic or survival function. Activated IGF-IR signals PI3K and downstream phosphorylation of Akt, or protein kinase B. Akt can effectively block, through phosphorylation, molecules such as BAD, which are essential for the initiation of programmed cell death, and inhibit initiation of apoptosis (Datta et al., Cell 91 :231-41 (1997)). Apoptosis is an important cellular mechanism that is critical to normal developmental processes (Oppenheim, Annu. Rev. Neurosci. 14:453-501 (1991)). It is a key mechanism for effecting the elimination of severely damaged cells and reducing the potential persistence of mutagenic lesions that may promote tumorigenesis. To this end, it has been demonstrated that activation of IGFs signaling can promote the formation of spontaneous tumors in a mouse transgenic model (DiGiovanni et al., Cancer Res. 60:1561-70 (2000)). Furthermore, IGF over-expression can rescue cells from chemotherapy induced cell death and may be an important factor in tumor cell drug resistance (Gooch et al., Breast Cancer Res. Treat. 56:1-10 (1999)). Consequently, modulation of the IGF signaling pathway has been shown to increase the sensitivity of tumor cells to chemotherapeutic agents (Benini et al., Clinical Cancer Res. 7:1790-97 (2001)). [0009] A large number of research and clinical studies have implicated the IGF-IR and its ligands (IGFs) in the development, maintenance, and progression of cancer. In tumor cells, over-expression of the receptor, often in concert with over-expression of IGF ligands, leads to potentiation of these signals and, as a result, enhanced cell proliferation and survival. IGF-I and IGF-II have been shown to be strong mitogens for a wide variety of cancer cell lines including prostate (Nickerson et al., Cancer Res. 61:6276-80 (2001); Hellawell et al., Cancer Res. 62:2942-50 (2002)) breast (Gooch et al., Breast Cancer Res. Treat. 56:1-10 (1999)), lung, colon (Hassan and Macaulay, Ann. Oncol. 13:349-56 (2002)), stomach, leukemia, pancreas, brain, myeloma (Ge and Rudikoff, Blood 96:2856-61 (2000), melanoma (All-Ericsson et al., Invest. Ophthalmol. Vis. Sci. 43:1-8 (2002)), and ovary (reviewed in: Macaulay, Br. J. Cancer 65:311-20 (1990)) and this effect is mediated through the IGF-IR. High circulating levels of IGF-I in serum have been associated with an increased risk of breast, prostate, and colon cancer (Pollak, Eur. J. Cancer 36:1224-28 (2000)). In a mouse model of colon cancer, increases in circulating IGF-I levels in vivo led to a significant increase in the incidence of tumor growth and metastasis (Wu et al., Cancer Res. 62: 1030-35 (2002)). Constitutive expression of IGF-I in epidermal basal cells of transgenic mice has been shown to promote spontaneous tumor formation (DiGiovanni et al., Cancer Res. 60:1561-1570 (2000; Bol et al., Oncogene 14:1725-1734 (1997)). Over-expression of IGF-II in cell lines and tumors occurs with high frequency and may result1 from loss of genomic imprinting of the IGF-II gene (Yaginuma et al., Oncology 54:502-7 (1997)). Receptor over- expression has been demonstrated in many diverse human tumor types including lung (Quinn et al., J. Biol. Chem. 271:11477-83 (1996)), breast (Cullen et al., Cancer Res. 50: 48-53 (1990); Peyrat and Bormeterre, Cancer Res. 22:59-67 (1992); Lee and Yee, Biomed. Pharmacother. 49:415-21 (1995)), sarcoma (van Nalen et al., J. Cancer Res. Clin. Oncol. 118:269-75 (1992); Scotlandi et al., Cancer Res. 56:4570-74 (1996)), prostate (Νickerson et al., Cancer Res. 61:6276-80 (2001)), and colon (Hassan and Macaulay, Ann. Oncol. 13:349- 56 (2002)). In addition, highly metastatic cancer cells have been shown to possess higher expression of IGF-II and IGF-IR than tumor cells that are less prone to metastasize (Guerra et al., Int. J. Cancer 65:812-20 (1996)). A critical role of the IGF-IR in cell proliferation and transformation was demonstrated in experiments of IGF-IR knockout derived mouse embryo fibroblasts. These primary cells grow at significantly reduced rates in culture medium containing 10% serum and fail to transform by a variety of oncogenes including SN40 Large T (Sell et al., Mol. Cell. Biol. 3604-12 (1994)). Recently it was demonstrated that resistance to the drug Herceptin in some forms of breast cancer may be due to activation of IGF-IR signaling in those cancers (Lu et al., J. Natl. Cancer Inst. 93:1852-57 (2001)). Over- expression or activation of IGF-IR may therefore not only be a major determinant in tumorigenicity, but also in tumor cell drug resistance. ( [0010] Activation of the IGF system has also been implicated in several pathological conditions besides cancer, including acromegaly (Drange and Melmed. In: The IGF System. Humana Press. 699-720 (1999)), retinal neovascularization (Smith et al., Nature Med. 12:1390-95 (1999)), and psoriasis (Wraight et al., Nature Biotech. 18:521-26 (2000)). In the latter study, an antisense oligonucleotide preparation targeting the IGF-IR was effective in significantly inhibiting the hyperproliferation of epidermal cells in human psoriatic skin grafts in a mouse model, suggesting that anti-IGF-IR therapies may be an effective treatment for this chronic disorder. [0011] A variety of strategies have been developed to inhibit the IGF-IR signaling pathway in cells. Antisense oligonucleotides have been effective in vitro and in experimental mouse models, as shown above for psoriasis. In addition, inhibitory peptides targeting the IGF-IR have been generated that possess anti-proliferative activity in vitro and in vivo (Pietrzkowski et al., Cancer Res. 52:6447-51 (1992); Haylor et al., J. Am. Soc. Nephrol. 11 :2027-35 (2000)). A synthetic peptide sequence from the C-terminus of IGF-IR has been shown to induce apoptosis and significantly inhibit tumor growth (Reiss et al., J Cell. Phys. 181 : 124-35 (1999)). Several dominant-negative mutants of the IGF-IR have also been generated which, upon over-expression in tumor cell lines, compete with wild-type IGF-IR for ligand and effectively inhibit tumor cell growth in vitro and in vivo (Scotlandi et al., Int. J. Cancer 101:11-6 (2002); Seely et al., BMC Cancer 2:15 (2002)). Additionally, a soluble form of the IGF-IR has also been demonstrated to inhibit tumor growth in vivo (D'Ambrosio et al., Cancer Res. 56:4013-20 (1996)). Antibodies directed against the human IGF-IR have also been shown to inhibit tumor cell proliferation in vitro and tumorigenesis in vivo including cell lines derived from breast cancer (Artega and Osborne, Cancer Res. 49:6237-41 (1989)), Ewing's osteosarcoma (Scotlandi et al., Cancer Res. 58:4127-31 (1998)), and melanoma (Furlanetto et al., Cancer Res. 53:2522-26 (1993)). Antibodies are attractive therapeutics chiefly because of they 1) can possess high selectivity for a particular protein antigen, 2) are capable of exhibiting high affinity binding to the antigen, 3) possess long half- lives in vivo, and, since they are natural immune products, should 4) exhibit low in vivo toxicity (Park and Smolen. In: Advances in Protein Chemistry. Academic Press. pp:360-421 (2001)). Antibodies derived from non-human sources, e.g.: mouse, may, however, effect a directed immune response against the therapeutic antibody, following repeated application, thereby neutralizing the antibody's effectiveness. Fully human antibodies offer the greatest potential for success as human therapeutics since they would likely be less immunogenic than murine or chimeric antibodies in humans, similar to naturally occurring immuno-responsive antibodies. To this end, there is a need to develop high affinity human anti-IGF-IR monoclonal antibodies for therapeutic use.
SUMMARY OF THE INVENTION [0012] The invention provides human monoclonal antibodies and fragments thereof that bind specifically to the human IGF-I receptor. The antibodies have at least one property selected from (i) inhibits binding of IGF-I or IGF-II to IGF-IR, (ii) neutralizes activation of IGF-IR by IGF-I or IGF-II, (iii) reduces IGF-IR surface receptor by at least about 80%; and (iv) binds to IGF-IR with a Kd of about 3 x 10"10 or less. In a more preferred embodiment, an antibody of the invention reduces IGF-IR surface receptor by at least about 85%, and more preferably by at least about 90%. Further, the antibodies inhibit ligand-mediated receptor autophosphorylation and downstream cellular signaling through the MAPK and Akt pathways. Antibodies of the invention, used alone or in combination with an anti-neoplastic agent, are particularly useful for treating neoplastic diseases and hyperprohferative disorders. [0013] The invention provides isolated polynucleotides encoding the antibodies or fragments thereof, expression vectors comprising the polynucleotide sequences, and host cells for expression. [0014] Further, the invention provides pharmaceutical compositions and diagnostic and therapeutic methods for treatment of tumors and hyperprohferative disease. The methods can further comprise administration of an anti-neoplastic agent or treatment.
BRIEF DESCRIPTION OF THE FIGURES [0015] Figure 1 depicts the nucleotide sequence of the 2F8 heavy chain variable domain. [0016] Figure 2 depicts the amino acid sequence of the 2F8 heavy chain variable domain. CDRs are in bold and underlined. [0017] Figure 3 depicts the nucleotide sequence of the complete 2F8 heavy chain (underline: secretory signal sequence; italics: IgGl constant region). [0018] Figure 4 depicts the amino acid sequence of the complete 2F8 heavy chain (underline: secretory signal sequence; bold: CDRs; italics: IgGl constant region). [0019] Figure 5 depicts the nucleotide sequence of the 2F8 light chain variable domain. [0020] Figure 6 depicts the amino acid sequence of the 2F8 light chain variable domain. CDRs are in bold and underlined. [0021] Figure 7 depicts the nucleotide sequence of the complete 2F8 light chain (underline: secretory signal sequence; italics: IgGl constant region). [0022] Figure 8 depicts the amino acid sequence of the complete 2F8 light chain (underline: secretory signal sequence; bold: CDRs; italics: IgGl constant region). [0023] Figure 9 depicts the nucleotide sequence of the A12 light chain variable domain. [0024] Figure 10 depicts the amino acid sequence of the A12 light chain variable domain. CDRs are in bold and underlined. [0025] Fig. 11 depicts the nucleotide sequence of the complete A12 light chain (underline: secretory signal sequence; italics: IgGl constant region). [0026] Figure 12 depicts the amino acid sequence of the complete A12 light chain (underline: secretory signal sequence; bold: CDRs; italics: IgGl constant region). [0027] Figure 13 depicts VH and NL CDR sequences of antibodies 2F8 and A12. Differences between the V CDRS are underlined. [0028] Figure 14 depicts the homology between 2F8 and A12 light chain variable region amino acid sequences. Sequences differences are boxed and CDRs are highlighted. [0029] Figure 15 shows results of an assay that measures the capacity of antibodies 2F8 and A12 to block binding of IGF-I to immobilized soluble IGF-IR. A12 antibodies having kappa or lambda light chain constant regions were tested. [0030] Figure 16 shows results of an assay that measures the capacity of antibodies 2F8 and A12 (lambda light chain) to block binding of IGF-I to MCF7 cells. [0031 ] Figure 17 shows results of an assay that measures the capacity of antibodies 2F8 and A12 to block insulin binding to ZR75-I cells. [0032] Figure 18 shows the effect of antibody A12 on 3H-thymidine incorporation in a mitogenesis assay. Figure 18 A: MCF7 breast cancer cells; Figure 18B: BxPC-3 pancreatic cancer cells; Figure 18C: HT-29 colon cancer cells. [0033] Figure 19 shows inhibition of IGF-I mediated receptor phosphorylation. Panel A: Inhibition in MCF7 breast cancer cells by antibodies A12'and 2F8. Panel B: Inhibition in HT-29 colorectal cancer cells and BxPC-3 pancreatic cancer cells by antibody A12. [0034] Figure 20 shows inhibition of IGF-I mediated phosphorylation of downstream effector molecules by antibodies A12 and 2F8. Panel A: inhibition of phosphorylation of MAPK; Panel B: inhibition of phosphorylation of Akt. [0035] Figure 21 shows binding of antibody A12 to human and murine IGF-IR positive and negative cell lines. MCF7: human breast cancer cells; R-: mouse embryo fibroblasts; HEL: human leukemia cells; Lewis Lung: mouse lung carcinoma cells. [0036] Figure 22 shows receptor internalization. Panel A shows internalization of labeled antibody A12 following binding to IGF-IR on MCF7 cells. Panel B shows depletion of cell-surface associated IGF-IR. Panel C shows degradation of total cellular IGF-IR after prolonged treatment with A12. [0037] Figure 23 shows inhibition of HT-29 human colon carcinoma growth in nude mice by antibody 2F8 and CPT-11 (irinotecan) alone or in combination. [0038] Figure 24 shows the effect of antibody A12 on HT-29 human colorectal tumor growth in nude mice. [0039] Figure 25 shows the effect of antibody A12 on MCF7 human breast cancer growth in nude mice. [0040] Figure 26 shows inhibition of BxPC-3 pancreatic cancer xenografts in nude mice by antibody A 12, gemcitabine, or CPT-11 (irinotecan) alone, or in combination. [0041] Figure 27 shows inhibition of HT-29 colorectal cancer xenografts in nude mice by antibody A12, paclitaxel, or CPT-11 (irinotecan) alone, or in combination.
DETAILED DESCRIPTION OF THE INVENTION [0042] The present invention provides antibodies and fragments thereof, specific for insulin-like growth factor-I receptor (IGF-IR), as well as isolated or purified polynucleotide sequences encoding the antibodies. In certain embodiments of the invention, human antibodies are provided. The antibodies can also be used in combination with other chemical and biological agents, including, but not limited to, anti-neoplastic agents and/or agents that are inhibitors of other receptors or receptor substrates mediating cell growth. The invention further relates to anti-neoplastic agents that are inhibitors of topoisomerase function. The choice of such agents is advantageous for use in therapeutic methods in combination with antibodies that are specific for IGF-IR. [0043] Naturally occurring antibodies typically have two identical heavy chains and two identical light chains, with each light chain covalently linked to a heavy chain by an interchain disulfide bond and multiple disulfide bonds further link the two heavy chains to one another. Individual chains can fold into domains having similar sizes (110-125 amino acids) and structures, but different functions. The light chain can comprise one variable domain (VL) and/or one constant domain (CL). The heavy chain can also comprise one variable domain (Nπ) and/or, depending on the class or isotype of antibody, three or four constant domains (CHI, CH2, CR3 and CR4). In humans, the isotypes are IgA, IgD, IgE, IgG, and IgM, with IgA and IgG further subdivided into subclasses or subtypes (IgA1-2 and [0044] Generally, the variable domains show considerable amino acid sequence variability from one antibody to the next, particularly at the location of the antigen-binding site. Three regions, called hypervariable or complementarity-determining regions (CDRs), are found in each of N and NH, which are supported by less variable regions called frameworks. [0045] The portion of an antibody consisting of N and NH domains is designated Fv (Fragment variable) and constitutes the antigen-binding site. Single chain Fv (scFv) is an antibody fragment containing a VL domain and a VH domain on one polypeptide chain, wherein the Ν terminus of one domain and the C terminus of the other domain are joined by a flexible linker (see, e.g., U.S. Pat. No. 4,946,778 (Ladner et al.); WO 88/09344, (Huston et al.). WO 92/01047 (McCafferty et al.) describes the display of scFv fragments on the surface of soluble recombinant genetic display packages, such as bacteriophage. [0046] The peptide linkers used to produce the single chain antibodies can be flexible peptides selected to assure that the proper three-dimensional folding and association of the VL and VH domains occurs. The linker is generally 10 to 50 amino acid residues. Preferably, the linker is 10 to 30 amino acid residues. More preferably the linker is 12 to 30 amino acid residues. Most preferably is a linker of 15 to 25 amino acid residues. An non-limiting example of such a linker peptides is (Gly-Gly-Gly-Gly-Ser)3 (SEQ ID NO:33). [0047] Fab (Fragment, antigen binding) refers to the fragments of the antibody consisting of VL-CL and VH-CH1 domains. Such a fragment generated by digestion of a whole antibody with papain does not retain the antibody hinge region by which two heavy chains are normally linked. The fragment is monovalent and simply referred to as Fab. Alternatively, digestion with pepsin results in a fragment that retains the hinge region. Such a fragment with intact interchain disulfide bonds linking two heavy chains is divalent and is referred to as F(ab')2- A monovalent Fab' results when the disulfide bonds of an F(ab')2 are reduced (and the heavy chains are separated. Because they are divalent, intact antibodies and F(ab')2 fragments have higher avidity for antigen that the monovalent Fab or Fab' fragments. WO 92/01047 (McCafferty et al.) describes the display of Fab fragments on the surface of soluble recombinant genetic display packages, such as bacteriophage. [0048] Fc (Fragment crystallization) is the designation for the portion or fragment of an antibody that consists of paired heavy chain constant domains, hi an IgG antibody, for example, the Fc consists of heavy chain CH2 and CH3 domains. The Fc of an IgA or an IgM antibody further comprises a CR4 domain. The Fc is associated with Fc receptor binding, activation of complement-mediated cytdtoxicity and antibody-dependent cellular-cytotoxicity (ADCC). For antibodies such as IgA and IgM, which are complexes of multiple IgG like proteins, complex formation requires Fc constant domains. [0049] Finally, the hinge region separates the Fab and Fc portions of the antibody, providing for mobility of Fabs relative to each other and relative to Fc, as well as including multiple disulfide bonds for covalent linkage of the two heavy chains. [0050] Antibody formats have been developed which retain binding specificity, but have other characteristics that may be desirable, including for example, bispecificity, multivalence (more than two binding sites), compact size (e.g., binding domains alone). [0051 ] Single chain antibodies lack some or all of the constant domains of the whole antibodies from which they are derived. Therefore, they can overcome some of the problems associated with the use of whole antibodies. For example, single-chain antibodies tend to be free of certain undesired interactions between heavy-chain constant regions and other biological molecules. Additionally, single-chain antibodies are considerably smaller than whole antibodies and can have greater permeability than whole antibodies, allowing single- chain antibodies to localize and bind to target antigen-binding sites more efficiently. Furthermore, the relatively small size of single-chain antibodies makes them less likely to provoke an unwanted immune response in a recipient than whole antibodies. [0052] Multiple single chain antibodies, each single chain having one VH and one VL domain covalently linked by a first peptide linker, can be covalently linked by at least one or more peptide linker to form a multivalent single chain antibodies, which can be monospecific or multispecific. Each chain of a multivalent single chain antibody includes a variable light chain fragment and a variable heavy chain fragment, and is linked by a peptide linker to at least one other chain. The peptide linker is composed of at least fifteen amino acid residues. The maximum number of amino acid residues is about one hundred. [0053] Two single chain antibodies can be combined to form a diabody, also known as a bivalent dimer. Diabodies have two chains and two binding sites, and can be monospecific or bispecific. Each chain of the diabody includes a VH domain connected to a VL domain. The domains are connected with linkers that are short enough to prevent pairing between domains on the same chain, thus driving the pairing between complementary domains on different chains to recreate the two antigen-binding sites. [0054] Three single chain antibodies can be combined to form triabodies, also known as trivalent trimers. Triabodies are constructed with the amino acid terminus of a VL or VH domain directly fused to the carboxyl terminus of a VL or VH domain, i.e., without any linker sequence. The triabody has three Fv heads with the polypeptides arranged in a cyclic, head- to-tail fashion. A possible conformation of the triabody is planar with the three binding sites located in a plane at an angle of 120 degrees from one another. Triabodies can be monospecific, bispecific or trispecific. [0055] Thus, antibodies of the invention and fragments thereof include, but are not limited to, naturally occurring antibodies, bivalent fragments such as (Fab')2, monovalent fragments such as Fab, single chain antibodies, single chain Fv (scFv), single domain antibodies, multivalent single chain antibodies, diabodies, triabodies, and the like that bind specifically with antigens. [0056] The antibodies of the present invention and particularly the variable domains thereof may be obtained by methods known in the art. These methods include, for example, the immunological method described by Kohler and Milstein, Nature, 256: 495-497 (1975) and Campbell, Monoclonal Antibody Technology, The Production and Characterization of Rodent and Human Hybridomas, Burdon et al., Eds., Laboratory Techniques in Biochemistry and Molecular Biology, Volume 13, Elsevier Science Publishers, Amsterdam (1985); as well as by the recombinant DNA methods such as described by Huse et al., Science, 246, 1275-81 (1989). The antibodies can also be obtained from phage display libraries bearing combinations of VH and VL domains in the form of scFv or Fab. The VH and VL domains can be encoded by nucleotides that are synthetic, partially synthetic, or naturally derived. In certain embodiments, phage display libraries bearing human antibody fragments can be preferred. Other sources of human antibodies are transgenic mice engineered to express human immunoglobulin genes. [0057] Antibody fragments can be produced by cleaving a whole antibody, or by expressing DNA that encodes the fragment. Fragments of antibodies may be prepared by methods described by Lamoyi et al., J. Immunol. Methods, 56: 235-243 (1983) and by Parham, J. Immunol. 131: 2895-2902 (1983). Such fragments may contain one or both Fab fragments or the F(ab')2 fragment. Such fragments may also contain single-chain fragment variable region antibodies, i.e. scFv, dibodies, or other antibody fragments. Methods of producing such functional equivalents are disclosed in PCT Application WO 93/21319, European Patent Application No. 239,400; PCT Application WO 89/09622; European Patent Application 338,745; and European Patent Application EP 332,424. [0058] The antibodies, or fragments thereof, of the present invention are specific for IGF-IR. Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Antibodies, or fragments thereof, of the present invention, for example, can be monospecific or bispecific. Bispecific antibodies (BsAbs) are antibodies that have two different antigen-binding specificities or sites. Where an antibody has more than one specificity, the recognized epitopes can be associated with a single antigen or with more than one antigen. Thus, the present invention provides bispecific antibodies, or fragments thereof, that bind to two different antigens, with at least one specificity for IGF-IR. [0059] Specificity of the present antibodies, or fragments thereof, for IGF-IR can be determined based on affinity and/or avidity. Affinity, represented by the equilibrium constant for the dissociation of an antigen with an antibody (Kd), measures the binding strength between an antigenic determinant and an antibody-binding site. Avidity is the measure of the strength of binding between an antibody with its antigen. Avidity is related to both the affinity between an epitope with its antigen binding site on the antibody, and the valence of the antibody, which refers to the number of antigen binding sites specific for a particular epitope. Antibodies typically bind with a dissociation constant (Kd) of IO"5 to 10"11 liters/mol. Any Kd greater than IO"4 liters/mol is generally considered to indicate nonspecific binding. The lesser the value of the Kd, the stronger the binding strength between an antigenic determinant and the antibody binding site. [0060] Antibodies of the present invention, or fragments thereof, also include those for which binding characteristics have been improved by direct mutation, methods of affimty maturation, phage display, or chain shuffling. Affinity and specificity can be modified or improved by mutating CDR and/or FW residues and screening for antigen binding sites having the desired characteristics (see, e.g., Yang et al., J. Mol. Biol., (1995) 254: 392-403). One way is to randomize individual residues or combinations of residues so that in a population of, otherwise identical antigen binding sites, subsets of from two to twenty amino acids are found at particular positions. Alternatively, mutations can be induced over a range of residues by error prone PCR methods (see, e.g., Hawkins et al., J. Mol. Biol, (1992) 226: 889-96). In another example, phage display vectors containing heavy and light chain variable region genes can be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol., (1996) 250: 359-68). These methods of mutagenesis are illustrative of the many methods known to one of skill in the art. [0061] Equivalents of the antibodies, or fragments thereof, of the present invention include polypeptides with amino acid sequences substantially the same as the amino acid sequence of the variable or hypervariable regions of the full-length anti-IGF-IR antibodies. Substantially the same amino acid sequence is defined herein as a sequence with at least 70%, preferably at least about 80%, and more preferably at least about 90% homology to another amino acid sequence, as determined by the FASTA search method in accordance with Pearson and Lipman (Proc. Natl. Acad. Sci. USA (1988) 85: 2444-8). [0062] Conservative amino acid substitution is defined as a change in the amino acid composition by way of changing one or two amino acids of a peptide, polypeptide or protein, or fragment thereof. The substitution is of amino acids with generally similar properties (e.g., acidic, basic, aromatic, size, positively or negatively charged, polarity, non-polarity) such that the substitutions do not substantially alter peptide, polypeptide or protein characteristics (e.g., charge, isoelectric point, affinity, avidity, conformation, solubility) or activity. Typical substitutions that may be performed for such conservative amino acid substitution may be among the groups of amino acids as follows: glycine (G), alanine (A), valine (V), leucine (L) and isoleucine (I); aspartic acid (D) and glutamic acid (E); alanine (A), serine (S) and threonine (T); histidine (H), lysine (K) and arginine (R): asparagine (N) and glutamine (Q); phenylalanine (F), tyrosine (Y) and tryptophan (W) [0063] Conservative amino acid substitutions can be made in, e.g., regions flanking the hypervariable regions primarily responsible for the selective and/or specific binding characteristics of the molecule, as well as other parts of the molecule, e.g., variable heavy chain cassette. [0064] Each domain of the antibodies of this invention can be a complete antibody with the heavy or light chain variable domain, or it can be a functional equivalent or a mutant or derivative of a naturally-occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Griffiths et al.). For instance, it is possible to join together domains corresponding to antibody variable domains, which are missing at least one amino acid. The important characterizing feature is the ability of each domain to associate with a complementary domain to form an antigen- binding site. Accordingly, the terms variable heavy and light chain fragment should not be construed to exclude variants that do not have a material effect on specificity. [0065] In a preferred embodiment, the anti-IGF-IR antibodies of the present invention are human antibodies that exhibit one or more of following properties. [0066] 1) The antibodies bind to the external domain of IGF-IR and inhibit binding of IGF-I or IGF-II to IGF-IR. Inhibition can be determined, for example, by a direct binding assay using purified or membrane bound receptor. In this embodiment, the antibodies of the present invention, or fragments thereof, preferably bind IGF-IR at least as strongly as the natural ligands of IGF-IR (IGF-I and IGF-II). [0067] 2) The antibodies neutralize IGF-IR. Binding of a ligand, e.g. , IGF-I or IGF- II, to an external, extracellular domain of IGF-IR stimulates autophosphorylation of the beta subunit and phosphorylation of IFG-IR substrates, including MAPK, Akt, and IRS-1. [0068] Neutralization of IGF-IR includes inhibition, diminution, inactivation and/or disruption of one or more of these activities normally associated with signal transduction. Further, this includes inhibition of IGF-IR / IR heterodimers as well as IGF-IR homodimers. Thus, neutralizing IGF-IR has various effects, including inhibition, diminution, inactivation and/or disruption of growth (proliferation and differentiation), angiogenesis (blood vessel recruitment, invasion, and metastasis), and cell motility and metastasis (cell adhesion and invasiveness). [0069] One measure of IGF-IR neutralization is inhibition of the tyrosine kinase activity of the receptor. Tyrosine kinase inhibition can be determined using well-known methods; for example, by measuring the autophosphorylation level of recombinant kinase receptor, and/or phosphorylation of natural or synthetic substrates. Thus, phosphorylation assays are useful in determining neutralizing antibodies in the context of the present invention. Phosphorylation can be detected, for example, using an antibody specific for phosphotyrosine in an ELISA assay or on a western blot. Some assays for tyrosine kinase activity are described in Panek et al., J. Pharmacol. Exp. Thera. 283: 1433-44 (1997) and Batley et al., Life Sci. 62:143-50 (1998). Antibodies of the invention cause a decrease in tyrosine phosphorylation of IGF-IR of at least about 75%, preferably at least about 85%, and more preferably at least about 90% in cells that respond to ligand. [0070] Another measure of IGF-IR neutralization is inhibition of phosphorylation of downstream substrates of IGF-IR. Accordingly, the level of phosphorylation of MAPK, Akt, or IRS-1 can be measured. The decrease in substrate phosphorylation is at least about 50%, preferably at least about 65%, more preferably at least about 80%. [0071] In addition, methods for detection of protein expression can be utilized to determine IGF-IR neutralization, wherein the proteins being measured are regulated by IGF- IR tyrosine kinase activity. These methods include immunohistochemistry (IHC) for detection of protein expression, fluorescence in situ hybridization (FISH) for detection of gene amplification, competitive radioligand binding assays, solid matrix blotting techniques, such as Northern and Southern blots, reverse transcriptase polymerase chain reaction (RT- PCR) and ELISA. See, e.g., Grandis et al., Cancer, 78:1284-92 (1996); Shimizu et al., Japan J. Cancer Res., 85:567-71 (1994); Sauter et al., _4m. J. Path., 148:1047-53 (1996); Collins, Glia 15:289-96 (1995); Radinsky et al., Clin. Cancer Res. 1:19-31 (1995); Petrides et al., Cancer Res. 50:3934-39 (1990); Hoffmann et al, Anticancer Res. 17:4419-26 (1997); Wikstrand et al, Cancer Res. 55:3140-48 (1995). [0072] In vivo assays can also be utilized to determine IGF-IR neutralization. For example, receptor tyrosine kinase inhibition can be observed by mitogenic assays using cell lines stimulated with receptor ligand in the presence and absence of inhibitor. For example, MCF7 (American Type Culture Collection (ATCC), Rockville, MD) stimulated with IGF-I or IGF-II can be used to assay IGF-IR inhibition. Another method involves testing for inhibition of growth of IGF-IR -expressing tumor cells or cells transfected to express IGF-IR. Inhibition can also be observed using tumor models, for example, human tumor cells injected into a mouse. [0073] The present invention is not limited by any particular mechanism of IGF-IR neutralization. The anti-IGF-IR antibodies of the present invention can bind externally to the IGF-IR cell surface receptor, block binding of ligand (e.g., IGF-I or IGF-II) and subsequent signal transduction mediated via the receptor-associated tyrosine kinase, and prevent phosphorylation of the IGF-IR and other downstream proteins in the signal transduction cascade. [0074] 3) The antibodies down modulate IGF-IR. The amount of IGF-IR present on the surface of a cell depends on receptor protein production, internalization, and degradation. The amount of IGF-IR present on the surface of a cell can be measured indirectly, by detecting internalization of the receptor or a molecule bound to the receptor. For example, receptor internalization can be measured by contacting cells that express IGF-IR with a labeled antibody. Membrane-bound antibody is then stripped, collected and counted. Internalized antibody is determined by lysing the cells and detecting label in the lysates., [0075] Another way is to directly measure the amount of the receptor present on the cell following treatment with an anti-IGF-IR antibody or other substance, for example, by fluorescence-activated cell-sorting analysis of cells stained for surface expression of IGF-IR. Stained cells are incubated at 37°C and fluorescence intensity measured over time. As a control, part of the stained population can be incubated at 4°C (conditions under which receptor internalization is halted). [0076] As described in the Examples, cell surface IGF-IR can be detected and measured using a different antibody that is specific for IGF-IR and that does not block or compete with binding of the antibody being tested. (Burtrum, et al. Cancer Res. 63:8912-21 (2003)) Treatment of an IGF-IR expressing cell with an antibody of the invention results in reduction of cell surface IGF-IR. In a preferred embodiment, the reduction is at least about 70%, more preferably at least about 80%, and even more preferably at least about 90% in response to treatment with an antibody of the invention. A significant decrease can be observed in as little as four hours. [0077] Another measure of down-modulation is reduction of the total receptor protein present in a cell, and reflects degradation of internal receptors. Accordingly, treatment of cells (particularly cancer cells) with antibodies of the invention results in a reduction in total cellular IGF-IR. In a preferred embodiment, the reduction is at least about 70%, more preferably at least about 80%, and even more preferably at least about 90%. [0078] The antibodies of the invention bind to IGF-IR with a Kd of about 3 x 10"10 M"1 or less, preferably about 1 x 10"10 M"1 or less, and more preferably about 3 x lO"11 E ar less. [0079] In an embodiment of the invention, the antibodies inhibit tumor growth. For example, subcutaneous xenograft tumors can be established by injection of cells of a cancer cell line into an immunodeficient mouse. The mice are then treated by intraperitoneal injection of antibodies, for example, every three days, and tumor size measured at regular intervals. Compared to control injections, antibodies of the invention inhibit tumor growth. In a preferred embodiment, an antibody of the invention promotes tumor regression when combined with an anti-neoplastic agent. Further, as exemplified below, in a more preferred embodiment, antibodies of the invention promoting tumor regression when used in a monotherapy. By promoting tumor regression is meant that administration of an effective amount of antibody, or an effective amount of a combination of an antibody and a neoplastic agent results in a reduction is size or necrosis of the tumor. In a preferred embodiment of the invention, tumor regression may be observed and continue for a period of at least about 20 days, more preferably at least about 40 days, more preferably at least about 60 days. Tumor regression may can be measured as an average across a group of subjects undergoing a particular treatment regimen, or can be measured by the number of subjects in a treatment group in which tumors regress. [0080] Preferred antibodies of the present invention, or fragments thereof, are human antibodies having one, two, three, four, five, and/or six complementarity determining regions (CDRs) selected from the group consisting of SEQ ID NO.14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28,and SEQ ID NO:30. Preferably, the antibodies (or fragments thereof) of the present invention have CDRs of SEQ ID NO:14, SEQ ID NO:16 and SEQ ID NO:18. Alternatively and also preferably, the present antibodies, or fragments thereof, have CDRs of SEQ ID NO:20, SEQ ID NO:22 and SEQ ID NO:24. Alternatively and also preferably, the present antibodies, or fragments thereof, have CDRs of SEQ ID NO:26, SEQ ID NO:28 and SEQ ID NO:30. The amino acid sequences of the CDRs are set forth below in Table 1.
TABLE 1 Heavy Chain (2F8/A12) CDR1 SYAIS SEQ ID NO: 14 CDR2 GΠPIFGTANYAQKFQG SEQ ID NO: 16
Figure imgf000019_0001
Light Chain (2F8) CDR1 QGDSLRSYYAS SEQ ID NO:20 CDR2 GKNNRPS SEQ ID NO:22 CDR3 NSRDNSDNRLI SEQ ID NO:24 Light Chain (A12) CDR1 QGDSLRSYYAT SEQ ID NO:26 CDR2 GENKRPS SEQ ID NO:28 CDR3 KSRDGSGQHLV SEQ ID NO:30
[0081] In another embodiment, the present antibodies, or fragments thereof, can have a heavy chain variable region of SEQ ID NO:l and/or a light chain variable region selected from SEQ ID NO:5 or SEQ ID NO:6. IMC-A12 is a particularly preferred antibody of the present invention. This antibody has human VH and VL framework regions (FWs) as well as CDRs. The VH variable domain of IMC-A12 (SEQ ID NO:l) has three CDRs corresponding to SEQ ID NOs:14, 16, and 18 and the VL domain (SEQ ID NO:5) has three CDRs corresponding to SEQ ID NOS:20, 22, and 24. IMC-2F8 is another preferred antibody of the present invention. This antibody also has human VH and VL framework regions (FWs) and CDRs. The VH variable domain of IMC-2F8 is identical to the VH variable domain of IMC- A12. The VL domain of IMC-2F8 (SEQ ID NO:9) has three CDRs corresponding to SEQ ID NOS:26, 28, and 30. [0082] hi another embodiment, antibodies of the invention compete for binding to IGF-IR with IMC-A12 and/or IMC-2F8. That is, the antibodies bind to the same or similar overlapping epitope. [0083] The present invention also provides isolated polynucleotides encoding the antibodies, or fragments thereof, described previously. The invention includes nucleic acids having a sequence encoding one, two, three, four, five and/or all six CDRs as set forth in Table 2. TABLE 2 Heavy Chain (2F8/A12) CDR1 agctatgcta tcagc SEQ ID NO: 13 CDR2 gggatcatcc ctatctttgg tacagcaaac tacgcacaga SEQ ID NO: 15 agttccaggg c CDR3 gcgccattac gatttttgga gtggtccacc caagaccact SEQ ID NO: 17 actactacta ctacatg gacgtc Light Chain (2F8) CDR1 caaggagaca gcctcagaag ctattatgca age SEQ ID NO: 19 CDR2 ggtaaaaaca accggccctc a SEQ ID N0:21 CDR3 aactcccggg acaacagtga taaccgtctg ata SEQ ID NO:23 Light Chain (A12) CDR1 caaggagaca gcctcagaag ctattatgca ace SEQ ID NO:25 CDR2 ggtgaaaata agcggccctc a SEQ ID NO:27 CDR3 aaatctcggg atggcagtgg tcaacatctg gtg SEQ ID NO:29
[0084] DNA encoding human antibodies can be prepared by recombining DNA encoding human constant regions and variable regions, other than the CDRs, derived substantially or exclusively from the corresponding human antibody regions and DNA encoding CDRs derived from a human (e.g., SEQ ID NOs:13, 15, and 17 for the heavy chain variable domain CDRs and SEQ ID NOs:19, 21, and 23 or SEQ ID NOS:25, 27 and 29 for the light chain variable domain CDRs). [0085] Other suitable sources of DNAs that encode fragments of antibodies include any cell, such as hybridomas and spleen cells, that express the full-length antibody. The fragments may be used by themselves as antibody equivalents, or may be recombined into equivalents, as described above. The DNA recombinations and other techniques described in this section may be carried out by known methods. Other sources of DNAs are single chain antibodies or Fabs produced from a phage display library, as is known in the art. [0086] Additionally, the present invention provides expression vectors containing the polynucleotide sequences previously described operably linked to an expression sequence, a promoter and an enhancer sequence. A variety of expression vectors for the efficient synthesis of antibody polypeptide in prokaryotic, such as bacteria and eukaryotic systems, including but not limited to yeast and mammalian cell culture systems have been developed. The vectors of the present invention can comprise segments of chromosomal, non- chromosomal and synthetic DNA sequences. [0087] Any suitable expression vector can be used. For example, prokaryotic cloning vectors include plasmids from E. coli, such as colEl , pCRl , pBR322, pMB9, pUC, pKSM, and RP4. Prokaryotic vectors also include derivatives of phage DNA such as MI 3 and other filamentous single-stranded DNA phages. An example of a vector useful in yeast is the 2μ plasmid. Suitable vectors for expression in mammalian cells include well-known derivatives of SV-40, adenovirus, retrovirus-derived DNA sequences and shuttle vectors derived from combination of functional mammalian vectors, such as those described above, and functional plasmids and phage DNA. [0088] Additional eukaryotic expression vectors are known in the art (e.g., PJ. Southern and P. Berg, J. Mol. Appl. Genet. lj. 327-41 (1982); Subramani et al., Mol. Cell. Biol. 1: 854-64 (1981); Kaufrnann and Sharp, "Amplification And Expression of Sequences Cotransfected with a Modular Dihydrofolate Reductase Complementary DNA Gene," J. Mol. Biol. 159: 601-21 (1982); Kaufrnann and Sharp, Mol. Cell. Biol. 159: 601-64 (1982); Scahill et al., "Expression And Characterization Of The Product Of A Human Immune Interferon DNA Gene hi Chinese Hamster Ovary Cells," Proc. Nat'lAcad. Sci. USA 80, 4654-59 (1983); Urlaub and Chasin, Proc. Nat'lAcad. Sci. USA 77: 4216-20, (1980). [0089] The expression vectors useful in the present invention contain at least one expression control sequence that is operatively linked to the DNA sequence or fragment to be expressed. The control sequence is inserted in the vector in order to control and to regulate the expression of the cloned DNA sequence. Examples of useful expression control sequences are the lac system, the trp system, the tac system, the trc system, major operator and promoter regions of phage lambda, the control region of fd coat protein, the glycolytic promoters of yeast, e.g., the promoter for 3-phosphoglycerate kinase, the promoters of yeast acid phosphatase, e.g., Pho5, the promoters of the yeast alpha-mating factors, and promoters derived from polyoma, adenovirus, retro virus, and simian virus, e.g., the early and late promoters or SV40, and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells and their viruses or combinations thereof. [0090] Where it is desired to express a gene construct in yeast, a suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7. Stinchcomb et al. Nature, 282: 39 (1979); Kingsman et al., Gene, 7: 141 (1979). The trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones, Genetics, 85: 12 (1977). The presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan. Similarly, Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene. [0091] The present invention also provides recombinant host cells containing the expression vectors previously described. Antibodies of the present invention can be expressed in cell lines other than in hybridomas. Nucleic acids, which comprise a sequence encoding a polypeptide according to the invention, can be used for transformation of a suitable mammalian host cell. [0092] Cell lines of particular preference are selected based on high level of expression, constitutive expression of protein of interest and minimal contamination from host proteins. Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines, such as but not limited to, COS-7 cells, Chinese Hamster Ovary (CHO) cells, Baby Hamster Kidney (BHK) cells and many others including cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells. Suitable additional eukaryotic cells include yeast and other fungi. Useful prokaryotic hosts include, for example, E. coli, such as E. coli SG-936, E. coli HB 101 , E. coli W3110, E. coli XI 776, E. coli X2282, E. coli DHI, and E. coli MRCl, Pseudomonas, Bacillus, such as Bacillus subtilis, and Streptomyces. [0093] These present recombinant host cells can be used to produce an antibody, or fragment thereof, by culturing the cells under conditions permitting expression of the antibody or fragment thereof and purifying the antibody or fragment thereof from the host cell or medium surrounding the host cell. Targeting of the expressed antibody or fragment for secretion in the recombinant host cells can be facilitated by inserting a signal or secretory leader peptide-encoding sequence (see, Shokri et al., Appl Microbiol Biotechnol. 60:654-64 (2003) Nielsen et al, Erot. Eng. 10:1-6 (1997) and von Heinje et al., Nucl. Acids Res. 14:4683-90 (1986)) at the 5' end of the antibody-encoding gene of interest. These secretory leader peptide elements can be derived from either prokaryotic or eukaryotic sequences. Accordingly suitably, secretory leader peptides are used, being amino acids joined to the N- terminal end of a polypeptide to direct movement of the polypeptide out of the host cell cytosol and secretion into the medium. [0094] The transformed host cells are cultured by methods known in the art in a liquid medium containing assimilable sources of carbon (carbohydrates such as glucose or lactose), nitrogen (amino acids, peptides, proteins or their degradation products such as peptones, ammonium salts or the like), and inorganic salts (sulfates, phosphates and/or carbonates of sodium, potassium, magnesium and calcium). The medium furthermore contains, for example, growth-promoting substances, such as trace elements, for example iron, zinc, manganese and the like. [0095] Another embodiment for the preparation of antibodies in the present invention is the expression of the nucleic acid encoding the antibody according to the invention in a transgenic animal that has a substantial portion of the human antibody producing genome inserted and is rendered deficient in the production of endogenous antibodies. Transgenic animals, include but not limited to mice, goat, and rabbit. One further embodiment of the invention, include expression of the antibody-coding gene in, for example, the mammary gland of the animal for secretion of the polypeptide during lactation. [0096] As described in the examples below, high affimty anti-IGF-IR antibodies according to the present invention can be isolated from a phage display library constructed from human heavy chain and light chain variable region genes. For example, a variable domain of the invention can be obtained from a peripheral blood lymphocyte that contains a rearranged variable region gene. Alternatively, variable domain portions, such as CDR and FW regions, can be derived from different human sequences. Over 90% of recovered clones after three rounds of selection are specific to IGF-IR. The binding affinities for IGF-IR of the screened Fabs can be in the nM range, which is as high as many bivalent anti- IGF-IR monoclonal antibodies produced using hybridoma technology. [0097] Antibodies, and fragments thereof, of the present invention can be obtained, for example, from naturally occurring antibodies, or Fab or scFv phage display libraries. Single domain antibodies can be obtained by selecting a VH or a VL domain from a naturally occurring antibody or hybridoma, or selected from a library of VH domains or a library of VL domains. It is understood that amino acid residues that are primary determinants of binding of single domain antibodies can be within Kabat defined CDRs, but may include other residues as well, such as, for example, residues that would otherwise be buried in the VH-VL interface of a VH-VL heterodimer. [0098] Antibodies of the present invention also include those for which binding characteristics have been improved by direct mutation, methods of affimty maturation, phage display, or chain shuffling. Affinity and specificity may be modified or improved by mutating CDRs and screening for antigen binding sites having the desired characteristics (see, e.g., Yang et al., J. Mol. Biol., 254: 392-403 (1995)). CDRs are mutated in a variety of ways. One way is to randomize individual residues or combinations of residues so that in a population of otherwise identical antigen binding sites, all twenty amino acids are found at particular positions. Alternatively, mutations are induced over a range of CDR residues by error prone PCR methods (see, e.g., Hawkins et al., J. Mol. Biol., 226: 889-896 (1992)). For example, phage display vectors containing heavy and light chain variable region genes may be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol., 250: 359-368 (1996)). These methods of mutagenesis are illustrative of the many methods known to one of skill in the art. [0099] The protein used to identify IGF-IR binding antibodies of the invention is preferably IGF-RI and, more preferably, is the extracellular domain of IGF-RI. The IGF-RI extracellular domain can be free or conjugated to another molecule. [0100] The antibodies of this invention can be fused to additional amino acid residues. Such amino acid residues can be a peptide tag, perhaps to facilitate isolation. Other amino acid residues for homing of the antibodies to specific organs or tissues are also contemplated. [0101] In another aspect of the invention, anti-IGF-IR antibodies or antibody fragments can be chemically or biosynthetically linked to anti-tumor agents or detectable signal-producing agents. As exemplified below, antibodies of the invention are efficiently internalized upon binding to cells bearing IGF-IR. Anti-tumor agents linked to an antibody include any agents which destroy or damage a tumor to which the antibody has bound or in the environment of the cell to which the antibody has bound. For example, an anti-tumor agent is a toxic agent such as a chemotherapeutic agent or a radioisotope. Suitable chemotherapeutic agents are known to those skilled in the art and include anthracyclines (e.g. daunomycin and doxorubicin), methotrexate, vindesine, neocarzinostatin, cis-platinum, chlorambucil, cytosine arabinoside, 5-fluorouridine, melphalan, ricin and calicheamicin. The chemotherapeutic agents are conjugated to the antibody using conventional methods (See, e.g., Hermentin and Seiler, Behringlnst. Mitt. 82:197-215(1988)). [0102] Detectable signal-producing agents are useful in vivo and in vitro for diagnostic purposes. The signal producing agent produces a measurable signal which is detectable by external means, usually the measurement of electromagnetic radiation. For the most part, the signal producing agent is an enzyme or chromophore, or emits light by fluorescence, phosphorescence or chemiluminescence. Chromophores include dyes which absorb light in the ultraviolet or visible region, and can be substrates or degradation products of enzyme catalyzed reactions. [0103] The invention further contemplates anti-IGF-IR antibodies or antibody fragments of the invention to which target or reporter moieties are linked. Target moieties are first members of binding pairs. Anti-tumor agents, for example, are conjugated to second members of such pairs and are thereby directed to the site where the antigen-binding protein is bound. A common example of such a binding pair is avidin and biotin. In a preferred embodiment, biotin is conjugated to an antigen-binding protein of the invention, and thereby provides a target for an anti-tumor agent or other moiety which is conjugated to avidin or streptavidin. Alternatively, biotin or another such moiety is linked to an antigen-binding protein of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin. [0104] Suitable radioisotopes for use as anti-tumor agents are also known to those skilled in the art. For example, I or At is used. These isotopes are attached to the antibody using conventional techniques (See, e.g., Pedley et al., Br. J. Cancer 68, 69- 73(1993)). Alternatively, the anti-tumor agent which is attached to the antibody is an enzyme which activates a prodrug. hi this way, a prodrug is administered which remains in its inactive form until it reaches the tumor site where it is converted to its cytotoxin form once the antibody complex is administered. In practice, the antibody-enzyme conjugate is administered to the patient and allowed to localize in the region of the tissue to be treated. The prodrug is then administered to the patient so that conversion to the cytotoxic drug occurs in the region of the tissue to be treated. Alternatively, the anti-tumor agent conjugated to the antibody is a cytokine such as interleukin-2 (IL-2), interleukin-4 (E -4) or tumor necrosis factor alpha (TNF-α). The antibody targets the cytokine to the tumor so that the cytokine mediates damage to or destruction of the tumor without affecting other tissues. The cytokine is fused to the antibody at the DNA level using conventional recombinant DNA techniques. [0105] A method of treating tumor growth in a mammal by administering to the mammal an effective amount of an antibody as previously described is also provided by the present invention. The IGF-IR signaling pathway has been extensively demonstrated to be a causative factor in the development of many types of cancer. IGF-I and IGF-II have been shown to be strong mitogens for a wide variety of cancer cell lines, including prostate, breast, colon, myeloma, ovary, pancreas and lung. Further, highly metastatic cancer cells have been shown to express higher levels of IGF-IR and IGF-II than tumor cells less prone to metastasize. [0106] Suitable tumors to be treated according to the present invention preferably express IGF-IR. While not intended to be bound to any particular mechanism, the diseases and conditions which can be treated or prevented by the present methods include, for example, those in which pathogenic angiogenesis or tumor growth is stimulated through an IGF-IR paracrine and/or autocrine loop. For example, highly metastatic tumors tend to express both IGF-II and IGF-IR. [0107] In an embodiment of the invention, anti-IGF-IR antibodies can be ' administered in combination with one or more other anti-neoplastic agents. For examples of combination therapies, see, e.g., U.S. Patent No. 6,217,866 (Schlessinger et al.) (Anti-EGFR antibodies in combination with anti-neoplastic agents); WO 99/60023 (Waksal et al.) (Anti- EGFR antibodies in combination with radiation). Any suitable anti-neoplastic agent can be used, such as a chemotherapeutic agent, radiation or combinations thereof. The anti- neoplastic agent can be an alkylating agent or an anti-metabolite. Examples of alkylating agents include, but are not limited to, cisplatin, cyclophosphamide, melphalan, and dacarbazine. Examples of anti-metabolites include, but not limited to, doxorubicin, daunorubicin, and paclitaxel, gemcitabine, and topoisomerase inhibitors irinotecan (CPT-11), aminocamptothecin, camptothecin, DX-8951f, and topotecan (topoisomerase I) and etoposide (VP-16) and teniposide (VM-26) (topoisomerase II). When the anti-neoplastic agent is radiation, the source of the radiation can be either external (external beam radiation therapy - EBRT) or internal (brachytherapy - BT) to the patient being treated. The dose of anti- neoplastic agent administered depends on numerous factors, including, for example, the type of agent, the type and severity tumor being treated and the route of administration of the agent. It should be emphasized, however, that the present invention is not limited to any particular dose. [0108] The anti-neoplastic agents which are presently known in the art or being evaluated can be grouped into a variety of classes including, for example, mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, anti survival agents, biological response modifiers, anti-hormones, and anti-angiogenesis agents. [0109] Among these classes, the data reported herein suggest that topoisomerase inhibitors are particularly effective anti-neoplastic agents when used in combination with antibodies that bind to IGF-IR. Accordingly, embodiments of the invention include methods in which a topoisomerase inhibitor is administered in combination with an antibody that binds to IGF-IR. The inhibitors can be inhibitors of topoisomerase I or topoisomerase II. Topoisomerase I inhibitors include irinotecan (CPT-11), aminocamptothecin, camptothecin, DX-8951f, topotecan. Topoisomerase II inhibitors include etoposide (VP-16), and teniposide (NM-26). Other substances are currently being evaluated with respect to topoisomerase inliibitory activity and effectiveness as anti-neoplastic agents. In a preferred embodiment, the topoisomerase inhibitor is irinotecan (CPT-11). The antibodies used in combination are antibodies of the invention that bind to IGF-IR and have at least one of the following properties: (i) inhibit binding of IGF-I or IGF-II to IGF-IR; (ii) neutralize activation of IGF- IR by IGF-I or IGF-II; (iii) reduce IGF-IR surface receptor; and bind to IGF-IR with a Kd of about 1 x 10"10 M"1 or less, hi a more preferred embodiment, the antibodies to be used in combination with a topoisomerase inhibitor have the characteristics of the human antibodies set forth above. [0110] Anti-IGF-IR antibodies of the invention can be admimstered with antibodies that neutralize other receptors involved in tumor growth or angiogenesis. In an embodiment of the invention, an anti-IGF-IR antibody is used in combination with a receptor antagonist that binds specifically to EGFR. Particularly preferred are antigen-binding proteins that bind to the extracellular domain of EGFR and block binding of one or more of its ligands and/or neutralize ligand-induced activation of EGFR. An EGFR antagonist can be an antibody that binds to EGFR or a ligand of EGFR and inhibits binding of EGFR to its ligand. Ligands for EGFR include, for example, EGF, TGF-α, amphiregulin, heparin-binding EGF (HB-EGF) and betacellulin. EGF and TGF-α are thought to be the main endogenous ligands that result in EGFR-mediated stimulation, although TGF-α has been shown to be more potent in promoting angiogenesis. It should be appreciated that the EGFR antagonist can bind externally to the extracellular portion of EGFR, which can or can not inhibit binding of the ligand, or internally to the tyrosine kinase domain. Examples of EGFR antagonists that bind EGFR include, without limitation, biological molecules, such as antibodies (and ftmctional equivalents thereof) specific for EGFR, and small molecules, such as synthetic kinase inhibitors that act directly on the cytoplasmic domain of EGFR. [0111] Another example of such a receptor is VEGFR. In an embodiment of the present invention, an anti-IGF-IR antibody is used in combination with a VEGFR antagonist, h one embodiment of the invention, an anti-IGF-IR antibody is used in combination with a receptor antagonist that binds specifically to VEGFR-1 /Fit- 1 receptor. In another embodiment, an anti-IGF-IR antibody is used in combination with a receptor antagonist that binds specifically to VEGFR-2/KDR receptor. Particularly preferred are antigen-binding proteins that bind to the extracellular domain of VEGFR-1 or VEGFR-2 and block binding by their ligands (VEGFR-2 is stimulated most strongly byVEGF; VEGFR-1 is stimulated most strongly by P1GF, but also by VEGF) and/or neutralize ligand-induced induced activation. For example, IMC-1121 is a human antibody that binds to and neutralizes VEGFR-2 (WO 03/075840; Zhu). Another example is MAb 6.12 is a scFv that binds to soluble and cell surface-expressed VEGFR-1. ScFv 6.12 comprises the V and VH domains of mouse monoclonal antibody MAb 6.12. A hybridoma cell line producing MAb 6.12 has been deposited as ATCC number PTA-3344 under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure and the regulations thereunder (Budapest Treaty). [0112] Other examples of growth factor receptors involved in tumorigenesis are the receptors for platelet-derived growth factor (PDGFR), nerve growth factor (NGFR), and fibroblast growth factor (FGFR). [0113] In an additional alternative embodiment, the IGF-IR antibody can be administered in combination with one or more suitable adjuvants, such as, for example, cytokines (IL-10 and IL-13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides. See, e.g., Larrivee et al, supra. It should be appreciated, however, that administration of only an anti- IGF-IR antibody is sufficient to prevent, inhibit, or reduce the progression of the tumor in a therapeutically effective manner. [0114] In a combination therapy, the anti-IGF-IR antibody is administered before, during, or after commencing therapy with another agent, as well as any combination thereof, i.e., before and during, before and after, during and after, or before, during and after commencing the anti-neoplastic agent therapy. For example, the anti-IGF-IR antibody can be administered between 1 and 30 days, preferably 3 and 20 days, more preferably between 5 and 12 days before commencing radiation therapy. In a preferred embodiment of the invention, chemotherapy is administered concurrently with or, more preferably, subsequent to antibody therapy. [0115] In the present invention, any suitable method or route can be used to administer anti-IGF-IR antibodies of the invention, and optionally, to co-administer anti- neoplastic agents and/or antagonists of other receptors. The anti-neoplastic agent regimens utilized according to the invention, include any regimen believed to be optimally suitable for the treatment of the patient's neoplastic condition. Different malignancies can require use of specific anti-tumor antibodies and specific anti-neoplastic agents, which will be determined on a patient to patient basis. Routes of administration include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration. The dose of antagonist administered depends on numerous factors, including, for example, the type of antagonists, the type and severity tumor being treated and the route of administration of the antagonists. It should be emphasized, however, that the present invention is not limited to any particular method or route of administration. [0116] It is noted that an anti-IGF-IR antibody of the invention can be administered as a conjugate, which binds specifically to the receptor and delivers a toxic, lethal payload following ligand-toxin internalization. The antibody-drug/small molecule conjugate can be directly linked to each other or via a linker, peptide or non-peptide. [0117] hi another aspect of the invention, an anti-IGF-IR antibody of the invention can be chemically or biosynthetically linked to one or more anti-neoplastic or anti-angiogenic agents. [0118] The invention further contemplates anti-IGF-IR antibodies to which target or reporter moieties are linked. Target moieties are first members of binding pairs. Anti- neoplastic agents, for example, are conjugated to second members of such pairs and are thereby directed to the site where the anti-IGF-IR antibody is bound. A common example of such a binding pair is avidin and biotin. In a preferred embodiment, biotin is conjugated to an anti-IGF-IR antibody, and thereby provides a target for an anti-neoplastic agent or other moiety, which is conjugated to avidin or streptavidin. Alternatively, biotin or another such moiety is linked to an anti-IGF-IR antibody of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin. [0119] It is understood that the anti-IGF-IR antibodies of the invention, where used in a mammal for the purpose of prophylaxis or treatment, will be administered in the form of a composition additionally comprising a pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins. The compositions of the injection can, as is well known in the art, be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the mammal. [0120] The present invention also includes kits for inhibiting tumor growth and/or angiogenesis comprising a therapeutically effective amount of a human anti-IGF-IR antibody. The kits can further contain any suitable antagonist of, for example, another growth factor receptor involved in tumorigenesis or angiogenesis (e.g., EGFR, VEGFR-1 /Fit- 1, VEGFR-2, PDGFR, NGFR, FGFR, etc, as described above). Alternatively, or in addition, the kits of the present invention can further comprise an anti-neoplastic agent. Examples of suitable anti- neoplastic agents in the context of the present invention have been described herein. The kits of the present invention can further comprise an adjuvant; examples have also been described above. [0121] Moreover, included within the scope of the present invention is use of the present antibodies in vivo and in vitro for investigative or diagnostic methods, which are well known in the art. The diagnostic methods include kits, which contain antibodies of the present invention. [0122] Accordingly, the present receptor antibodies thus can be used in vivo and in vitro for investigative, diagnostic, prophylactic, or treatment methods, which are well known in the art. Of course, it is to be understood and expected that variations in the principles of invention herein disclosed can be made by one skilled in the art and it is intended that such modifications are to be included within the scope of the present invention.
EXAMPLES [0123] The following examples further illustrate the invention, but should not be construed to limit the scope of the invention in any way. Detailed descriptions of conventional methods, such as those employed in the construction of vectors and plasmids, the insertion of genes encoding polypeptides into such vectors and plasmids, the introduction of plasmids into host cells, and the expression and determination thereof of genes and gene products can be obtained from numerous publications, including Sambrook, J et al., (1989) Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press; and Coligan, J. et al. (1994) Current Protocols in Immunology, Wiley & Sons, Incorporated All references mentioned herein are incorporated in their entirety.
Selection and engineering of anti-human IGF-IR monoclonal antibodies. [0124] In order to isolate high affinity antibodies to the human IGF-I receptor, recombinant extracellular portion of human IGF-IR was used to screen a human naive (non- immunized) bacteriophage Fab library containing 3.7xl010 unique clones (de Haard et al., J. Biol. Chem. 274:18218-30 (1999)). Soluble IGF-IR (50μg/ml) was coated onto tubes and blocked with 3% milk/PBS at 37 degrees for 1 hour. Phage were prepared by growing library stock to log phase culture, rescuing with M13K07 helper phage, and amplifying overnight at 30°C in 2YTAK culture medium at containing ampicillin and kanamycin selection. The resulting phage preparation was precipitated in 4% PEG/0.5M NaCI and resuspended in 3% milk/PBS. The immobilized receptors were then incubated with phage preparation for 1 hour at room temperature. Afterwards, the tubes were washed 10 times with PBST (PBS containing 0.1% Tween-20) followed by 10 times with PBS. The bound phage were eluted at RT for 10 min with 1 ml of a freshly prepared solution of 100 mM triethylamine. The eluted phage were incubated with 10 ml of mid-log phase TGI cells at 37°C for 30 min stationary and 30 min shaking. The infected TGI cells were pelleted and plated onto several large 2YTAG plates and incubated overnight at 30°C. All colonies that grew on the plates were scraped into 3 to 5 ml of 2YTA medium, mixed with glycerol (final concentration: 10%), aliquoted and stored at -70°C. For second round selection, 100 μl of the phage stock was added to 25 ml of 2YTAG medium and grown to mid-log phase. The culture was rescued with M13K07 helper phage, amplified, precipitated, and used for selection following the procedure described above, but with reduced concentration (5μg/ml) of IGF-IR immobilized onto tubes and increasing the numbers of washes following the binding process. A total of two rounds of selection were performed. [0125] Individual TGI clones were picked and grown at 37°C in 96 well plates and rescued with M13K07 helper phage as described above. The amplified phage preparation was blocked with 1/6 volume of 18% milk/PBS at RT for 1 h and added to Maxi-sorb 96-well microtiter plates (Nunc) coated with IGF-IR (1 μg/ml x 100 μl). After incubation at RT for 1 h the plates were washed 3 times with PBST and incubated with a mouse anti-M13 phage- HRP conjugate (Amersham Pharmacia Biotech, Piscataway, NJ). The plates were washed 5 times, TMB peroxidase substrate (KPL, Gaithersburg, MD) added, and the absorbance at 450 nm read using a microplate reader (Molecular Device, Sunnyvale, CA). From 2 rounds of selection, 80% of independent clones were positive for binding to IGF-IR. [0126] The diversity of the anti-IGF-IR Fab clones after the second round of selection was analyzed by restriction enzyme digestion pattern (i.e., DNA fingerprint). The Fab gene insert of individual clones was PCR amplified using primers: PUC19 reverse (5'- AGCGGATAACAATTTCACACAGG-3'; SEQ ID NO:31) and fdtet seq (5'- GTCGTCTTTCCAGACGTTAGT-3'; SEQ ID NO:32) which are specific for sequences flanking the unique Fab gene regions within the phage vector. Each amplified product was digested with a frequent-cutting enzyme, BstN I, and analyzed on a 3% agarose gel. A total of 25 distinct patterns were identified. DNA sequences of representative clones from each digestion pattern were determined by dideoxynucleotide sequencing. [0127] Plasmids from individual clones exhibiting positive binding to IGF-IR and unique DNA profile were used to transform a nonsuppressor E.coli host HB2151. Expression of the Fab fragments in HB2151 was induced by culturing the cells in 2YTA medium containing 1 mM isopropyl- 1-thio-β-D-galactopyranoside (IPTG, Sigma) at 30°C. A periplasmic extract of the cells was prepared by resuspending the cell pellet in 25 mM Tris (pH 7.5) containing 20% (w/v) sucrose, 200 mM NaCI, 1 mM EDTA and 0.1 mM PMSF, followed by incubation at 4°C with gentle shaking for 1 h. After centrifugation at 15,000 rpm for 15 min, the soluble Fab protein was purified from the supernatant by affinity chromatography using Protein G column followed the manufacturer's protocol (Amersham Pharmacia Biotech). [0128] Candidate binding Fab clones were screened for competitive blocking of radiolabeled human IGF-I ligand to immobilized IGF-IR (lOOng/well) coated onto 96 strip- well plates. Fab preparations were diluted and incubated with IGF-IR plates for 0.5-1 hour at room temperature in PBS/0.1%BSA. Forty 40 pM of 125I-IGF-I was then added and the plates incubated an additional 90 minutes. Wells were then washed 3 times with ice-cold PBS/0.1% BSA, dried, and then counted in a gamma scintillation counter. Candidates that exhibited greater than 30% inhibition of control radiolabeled ligand binding in single point assay were selected and in vitro blocking titers determined. Four clones were identified. Of these, only Fab clone 2F8 was shown to inhibit ligand binding by more than 50%, with an IC50 of approximately 200nM, and it was selected for conversion to full length IgGl format. The heavy chain variable region sequence and translated amino acid sequence for 2F8 is shown in Figures 1 and 2, respectively. The DNA sequence and translated polypeptide sequence of the 2F8 heavy chain engineered as full length IgGl are shown in Figures 3 and 4, respectively. [0129] Fab 2F8 sequencing determined that this Fab possessed a lambda light chain constant region. The DNA sequence and translated amino acid sequence of the 2F8 light chain are shown in Figures 5 and 6, respectively. The sequences for full-length lambda light chain format are shown in Figures 7 and 8. Binding kinetic analysis was performed on 2F8 IgG using a BIAcore unit. This antibody was determined to bind to the IGF-IR with an affinity of 0.5 -1 nM (0.5-1 x IO"9 M). [0130] In order to improve the affinity of this antibody, a second generation Fab phage library was generated in which the 2F8 heavy chain was conserved and the light chain was varied to a diversity of greater than 10 unique species. This method is termed light chain shuffling and has been used successfully to affinity mature selected antibodies for a given target antigen (Chames et al., J. Immunol. 169:1110-18 (2002)). This library was then screened for binding to the human IGF-IR (10 μg/ml) following procedures as described above, and the panning process repeated an additional three rounds with reduced IGF-IR concentration (2 μg/ml) for enrichment of high affinity binding Fabs. Seven clones were analyzed following round four. All 7 contained the same DNA sequence and restriction digest profile. The single isolated Fab was designated A12 and shown to possess a lambda light chain constant region. The light chain DNA sequence is shown in Figure 9 and amino acid sequence in Figure 10. Complete lambda light chain sequence and translated polypeptide sequence are shown in Figures 11 and 12, respectively. Amino acid sequence comparison of 2F8 and A12 light chains determined that the two variable regions differed by a total of 11 amino acids (refer to Figures 13 and 14). Nine of the changes were present within CDR regions, with the majority (6 amino acid residues) occurring within CDR3. [0131] A comparison of the two antibody (full IgG) affinities for human IGF-IR and their ligand blocking activity is shown in Table 3. Binding results were determined by human IGF-IR ELISA and represent the concentration of titered antibody necessary to achieve 50% binding relative to saturation. Blocking results represent the level of antibody necessary to inhibit 50% binding of 1 *ϊ I-IGF-I ligand to immobilized human IGF-IR. Affinity was determined by BIAcore analysis according to manufacturer's specifications (Pharmacia BIACORE 3000). Soluble IGF-IR was immobilized on the sensor chips and antibody binding kinetics determined.
Figure imgf000034_0001
[0132] The antibody changes incurred in 2F8 light chain to generate antibody A12 effected a significantly higher affinity of A12 for IGF-IR than 2F8. Concomitantly, this increase effected a greater binding ability of A12 for the receptor, as determined by ELISA, and at least a three-fold increase in blocking activity of ligand for immobilized receptor. Figure 15 shows a representative titration of the two anti-IGF-IR antibodies in receptor blocking assay. The activity of A12 remained the same, irrespective of whether the light chain was engineered with a human lambda or kappa class constant region. Antibody A12 engineered with a lambda class light chain was utilized in all subsequent procedures. In this assay, A12 inhibited the binding of radiolabeled IGF-I to IGF-IR to a greater extent than competition with cold ligand. The activity of 2F8 was comparable to competition with cold ligand. This is consistent with the relative affinities of the two antibodies (see Table 3) and IGF-I (0.5-1 nM).
Engineering and expression of fully human IsGl anti-IGF-IR antibodies from Fab clones. [0133] The DNA sequences encoding the heavy and light chain genes of Fabs 2F8 and A12 were amplified by polymerase chain reaction (PCR) using the Boerhinger Mannheim Expand kit according to manufacturer's instructions. Forward and reverse primers contained sequences for restriction endonuclease sites for cloning into mammalian expression vectors. The recipient vector for the heavy chain contained the entire human gamma 1 constant region cDNA sequence, flanked by a strong eukaryotic promoter and a 3 ' polyadenylation sequence. The full-length lambda light chain sequences for 2F8 or A12 were each cloned in to a second vector possessing only the eukaryotic regulatory elements for expression in mammalian cells. A selectable marker was also present on this vector for selection of stable DNA integrants following transfection of the plasmid into mammalian cells. Forward primers were also engineered with sequences encoding a strong mammalian signal peptide sequence for proper secretion of the expressed antibody. Following identification of properly cloned immunoglobulin gene sequences, the DNAs were sequenced and tested for expression in transient transfection. Transient transfection was performed into the COS7 primate cell line using Lipofection, according to manufacturer's specifications. At 24 or 48 hours post-transfection, the expression of full IgG antibody was detected in conditioned culture supernatant by anti-human-Fc binding ELISA. ELISA Plates (96 well) were prepared by coating with 100 ng/well of a goat-anti-human Fc-specific polyclonal antibody (Sigma) and blocked with 5% milk/PBS overnight at 4°C. The plates were then washed 5 times with PBS. Conditioned supernatant was added tol wells and incubated for 1.5 hours at room temperature. Bound antibody was detected with a goat anti-human lambda light chain-HRP antibody (Sigma) and visualized with TMB reagents and microplate reader as described above. Large scale preparation of anti-IGF-IR antibodies was achieved by either large scale transient transfection into COS cells, by scale-up of the Lipofection method or by stable transfection into a suitable host cell such as a mouse myeloma cell line (NSO, Sp2/0) or a Chinese hamster ovary cell line (CHO). Plasmid encoding the anti-IGF-IR antibodies were transfected into host cells by electroporation and selected in appropriate drug selection medium for approximately two weeks. Stably selected colonies were screened for antibody expression by anti-Fc ELISA and positive clones expanded into serum free cell culture medium. Antibody production from stably transfected cells was performed in suspension culture in spinner flasks or bioreactors for a period of up to two weeks. Antibody generated by either transient or stable transfection was purified by ProA affinity chromatography (Harlow and Lane. Antibodies. A Laboratory Manual. Cold Spring Harbor Press. 1988), eluted into a neutral buffered saline solution, and quantitated.
Determination of ligand blocking activity of anti-IGF-IR monoclonal antibodies on human tumor cells. [0134] The anti-IGF-IR antibodies were then tested for blocking of radiolabeled ligand to native IGF-IR on human tumor cells. Assay conditions were performed according to Arteaga and Osborne (Cancer Res. 49:6237-41 (1989)), with minor modifications. MCF7 human breast cancer cells were seeded into 24 well dishes, and cultured overnight. Sub- confluent monolayers were washed 2-3 times in binding buffer (Iscove's Medium/0.1% BSA) and antibody added in binding buffer. After a short incubation with the antibody at room 1 temperature, 40 pM I-IGF-I (approximately 40,000 cpm/well) was added to each well and incubated for an additional hour with gentle agitation. The wells were then washed three times with ice-cold PBS / 0.1% BSA. Monolayers were then lysed with 200 μl 0.5N NaOH and counted in a gamma counter. The results are shown graphically in Figure 16. On human tumor cells, antibody A12 inhibited ligand binding to IGF-IR with an IC5o of 3 nM (0.45 μg/ml). This was slightly lower than the inhibitory activity of cold IGF-I ligand (IC50 = 1 nM), but better than the inhibitory activity of cold IGF-II (IC5o = 9 nM). The differences observed for the two IGF ligands can likely be attributed to the slower binding kinetics of IGF-II for the IGF-IR than ligand IGF-I (Jansson et al., J Biol. Chem. 272:8189-97 (1997). The IC50 for antibody 2F8 was determined to be 30 nM (4.5 μg/ml). We subsequently determined the IGF-I ligand blocking activity of the A12 antibody on several different human tumor types. The results are shown in Table 4. Antibody A12 was effective in binding to endogenous cellular IGF-IR and inhibiting ligand binding to a range of human tumor types including cell lines from breast, pancreatic, and colorectal tissue.
Figure imgf000037_0001
[0135] The IGF-IR shares considerable homology with the insulin receptor (IR). To determine if the anti-IGF-IR antibodies were specific to this IGF-IR and did not block insulin binding, a cell-based blocking assay was performed on human ZR-75I breast cancer cells. Because insulin can bind to IGF-IR, albeit at three orders of magnitude lower affinity than for the IR, we utilized the human breast cancer line ZR-75I that possesses a higher IR to IGF-IR ratio in comparison to MCF7 cells. By using this line, we reasoned that insulin binding to the cells would be more indicative of specific IR binding. The assay was performed as described above for MCF7 cells and the results shown in Figure 17. Although cold insulin was able to titrate the binding of radiolabeled insulin to cells, neither 2F8 nor the high affinity A12 antibody blocked insulin binding, even at a concentration of 200 nM antibody, consistent with selective binding of these antibodies to IGF-IR and not IR.
Antibody-mediated inhibition oflisand-dependent cell mitogenesis. [0136] In order to determine if blocking of IGF-I binding to IGF-IR inhibited cellular proliferation, a mitogenic assay was performed on MCF7 breast cancer cells, BxPC-3 pancreatic cancer cells and HT-29 colon cancer cells. The assay was performed according to Prager, et al. (Proc. Natl. Acad. Sci. U.S.A. 91:2181-85 (1994), with some modification. Cells were plated into 96-well tissue culture plates at 5000-10000 cells/well and allowed to adhere overnight. The medium was then replaced with serum free defined medium and incubated overnight at 37°C. Cells were then incubated with IGF-I with or without antibody A12 and incubated overnight at 37°C. 0.25 μCi [ H]thymidine was then added to each well and incubated for 5 hours at 37°C. The supernatant was aspirated and the cells suspended by trypsinization for 5 minutes. The cells were then collected onto a filter and washed three times with water, using a cell harvester. After drying, the filter was processed for reading in a scintillation counter. The results are shown in Figures 18 A, B and C (MCF7, BxPC-3 and HT-29 respectively). IGF-I datapoints show titration of the ligand to determine the amount necessary to achieve the maximum mitogenic response. In measuring the activity of antibody A12 on various cancer cell types, IGF-I was added at a concentration of 5 nM and the antibody titered from 200 nM to 0.05 nM. Antibody A12 inhibited MCF7 mitogenesis in response to IGF-I ligand in a dose-dependent fashion, with an IC50 of 6 nM. [0137] Antibody A12 was then tested for mitogenic inhibition on several additional human tumor cells lines and the results shown in Table 5. Antibody A12 was effective at inhibiting IGF-I ligand-mediated mitogenesis of a variety of human tumor cell lines, including breast cancer, colorectal cancer, and multiple myeloma.
Figure imgf000038_0001
Antibody-mediated inhibition of IGF-I directed receptor phosphorylation and downstream signalins. [0138] To visualize the inhibitory effect of the anti-IGF-IR antibodies on IGF-I signaling, receptor auto-phosphorylation and downstream effector molecule phosphorylation analysis was performed in the presence or absence of antibody A12 or 2F8. The MCF7 human breast cancer cell line was selected for use due to its high IGF-IR density. Cells were plated into 10 cm or 6 well culture dishes and grown to 70-80% confluence. The monolayers were then washed twice in PBS and cultured overnight in serum free defined medium. Anti- IGF-IR antibody was then added in fresh serum-free media (100 nM-10 nM) and incubated cells 30 minutes before addition of ligand (10 nM). Cells were incubated with ligand for 10 minutes, then placed on ice and washed with ice-cold PBS. The cells were lysed by the addition of lysis solution (50 mM Tris-HCl, pH 7.4, 150 mM NaCI, 1% TritonX-100, 1 mM EDTA, 1 mM PMSF, 0.5 mM Na3VO4, 1 μg/ml leupeptin, 1 μg/ml pepstatin, and 1 μg/ml aprotinin) and the cells scraped into a centrifuge tube kept on ice for 15 minutes. The lysate was then clarified by centrifugation at 4°C. Solubilized IGF-IR was then immunoprecipitated (IP) from the lysate. Antibody 3B7 (Santa Cruz) or A12 at 1 μg/ml were incubated with 400 μl of lysate overnight at 4°C. Immune complexes were then precipitated by the addition of ProA-sepharose beads for 2 hours at 4°C, pelleted, and washed 3 times with lysis buffer. IP s bound to the ProA beads were stripped into denaturing gel running buffer. Lysate or IP were processed for denaturing gel electrophoresis and run on a 4-12% acrylamide gel and blotted to nylon or nitrocellulose membrane by western blot according to Towbin et al. (Biotechnology 24: 145-9 (1992)). Tyrosine phosphorylated protein was detected on the blot using an anti-p-tyrosine antibody (Cell Signaling #9411) and an anti-mouse-HRP secondary antibody. IGF-IR was detected with monoclonal antibody C-20 (Santa Cruz). For Akt phosphorylation, phospho-Akt was detected with antibody #559029 and total Akt with #559028 (BD Pharmingen). For MAPK phosphorylation, phospho-p44/42 was detected with #9101 and total p44/42 with #9102 (Cell Signaling Tech.). Bands were visualized with the ECL reagent on X-ray film. [0139] As shown in Figure 19 A, auto-phosphorylation of the IGF-IR in MCF7 cells was arrested following serum deprivation, and the addition of either 2F8 or A12 alone did not induce receptor phosphorylation, thereby demonstrating a lack of detectable agonist activity. Upon the addition of 10 nM IGF-I, IGF-IR phosphorylation was strongly induced. Antibody 2F8 effected an approximately 50% reduction in IGF-IR phosphorylation, whereas the high affinity antibody A12 nearly completely blocked phosphorylation. Similarly, antibody A12 inhibited auto-phosphorylation of IGF-IR in HT-29 colorectal and BxPC-3 pancreatic cancer cells (Fig. 19B). [0140] Downstream effector signaling in response to IGF-I was also inhibited by the anti-IGF-IR antibodies (Fig. 20). MAPK phosphorylation was considerably inhibited by both 2F8 and A12. Phosphorylation of the anti-apoptotic molecule Akt was less sensitive to anti- IGF-IR antibody blockade with 2F8. It effected only a slight reduction in Akt phosphorylation. A12 significantly inhibited Akt phosphorylation, even at a concentration of lOnM. Antibody A12 was equally proficient in immunoprecipitating solubilized IGF-IR as the commercial antibody 3B7, but A12 was not capable of detecting denatured IGF-IR immobilized on nylon membranes following western blot transfer.
FACs binding analysis of monoclonal antibody Al 2 to tumor cell lines. [0141] Since A12 was capable of immunoprecipitating endogenous IGF-IR, we were therefore interested in determining if A12 could also be used as detection antibody for fluorescence activated cell sorting (FACs). Human tumor cell lines were grown in culture, scraped into ice-cold PBS, and counted. Primary antibody, A12 (0.5μg), was added to approximately 5 million cells in 250 μl PBS/5%FBS and incubated on ice for 1 hour. The cells were then diluted to 3 mis in PBS/5% FBS, pelleted, and the supernatant aspirated. Secondary phycoerythin (PE) -labeled goat anti-human IgG F(ab)2 fragment was then added in 250 μl PBS/5% FBS at 1 :200 and incubated on ice for 60 minutes. Afterwards, the cells were again diluted and pelleted, as before, then resuspended in 500 μl PBS/5% FBS. FACs analysis was then performed on a Epics XL unit (Coulter). As shown in Fig. 21, antibody A12 fully shifted the human breast cancer cell line MCF7 and the human leukemia cell line HEL. IGF-IR negative mouse embryo fibroblasts (R- cells) (obtained from R. Baserga, Thomas Jefferson University, Philadelphia, PA) served as the negative control. A12 failed to bind to these cells, indicative of antibody binding specificity for the IGF-IR. A12 did, however, bind and partially shift the mouse tumor cell line Lewis Lung carcinoma, suggesting that this anti-human IGF-IR antibody possesses some cross-reactivity for the mouse IGF-IR.
IGF-I receptor internalization following binding of antibody A12. [0142] Antibody A12 has been shown to bind native IGF-IR on human tumor cells with high affinity. Antibody A12 was radio-iodinated with 125iodine using IODO-beads (Pierce) according to manufacturer's instructions. MCF7 human breast cancer cells were plated into 6-well plates and cultured overnight to 50% confluence. One microgram of 125I- A12 was added to each well and incubated at 37°C or kept on ice at 4°C. Plates were incubated for 30 minutes, 90 minutes, or 180 minutes and each time point performed in triplicate. The culture at 4°C was held for 180 minutes. At each time point, wells were washed lx w/PBS, then stripped for 5 minutes with lOOmM glycine-HCl, 2M urea, pH 2.5. The stripped material, representing membrane bound antibody was kept on ice for counting. Wells were then washed 3 times with PBS and cells solubilized with IN NaOH/1% TritonXlOO. The solubilized fraction represented the internalized antibody. Stripped and solublized fractions were then read on a gamma counter and plotted with standard deviation. As shown in Fig. 22, the level of internalized radioactivity increased with time in the cells cultured at 37°C, while little uptake was observed in cells maintained at 4°C where membrane transport should be severely retarded. This demonstrated that, upon binding to the IGF-IR, antibody A12 is rapidly internalized, potentially leading to a depletion of surface bound receptor. [0143] IGF-IR surface receptor density was determined by FACS. Adherent MCF 7 cells were treated for 4 h with 50 nM of antibody A12 of IGF-I at 37°C. Cells were washed in ice-cold PBS/5% BSA twice, and 1 x IO6 cells were aliquoted to staining tubes and placed on ice. An IGF-IR alpha-polypeptide specific, non-blocking mouse monoclonal antibody (Ab-1, NeoMarkers, Fremont, CA), was then incubated with cells at 4°C for 2 h. After PBS/BSA washes, cells were incubated with anti-mouse IgG phycoerythrin-conjugated secondary antibody (PharMingen, BD Biosciences) for 1 h on ice. After PBS/BSA wash, cells were analyzed by fluorescence-activated cell-sorting assay using a FACSvantage SE flow cytometer (BD Bioscience). As shown in Fig. 22b, exposure to A12 resulted in a significant reduction in the fluorescence intensity of MCF7 cells, indicative of surface receptor down-modulation due to internalization. Calculated mean fluorescence intensity ratio indicated a reduction in IGF-IR surface staining of 90% after incubation with A12. This shift was not seen when cells were incubated with A12 at 4°C, consistent with an energy- dependent, antibody-mediated, receptor internalization process. Exposure of cells to IGF-I did not cause a significant change in surface IGF-IR fluorescence intensity, consistent with Western blot analysis showing little effect of ligand on IGF-IR degradation [0144] Total cellular IGF-IR was determined HT-29 cells and BxPC-3 cells in response to treatment with IGF-I or A12. As shown in Fig. 22c, addition of IGF-I to growing cultures had no effect on IGF-IR expression. In contrast, addition of antibody A12 resulted in severe depletion of IGF-IR levels in the cells after 3-6 hours. Growth inhibition of human colorectal tumors alone or in combination with irinotecan (CPT- 11). [0145] We were interested to determine if the anti-IGF-IR antibodies were capable of inhibiting human tumor growth in vivo in a nude mouse xenograft model. Tumors were induced in 3-4 week old athymic nude (nu/nu) mice by subcutaneous injection of 2-3 million viable HT-29 human colorectal cancer cells in cell culture medium. The tumors were allowed to establish and antibody treatment started when the tumor volume reached 200 mm3. Ten animals were injected with tumor cells per treatment group. Antibody was injected intraperitoneally (IP) every three days at 1 mg or 0.5 mg in 0.5 ml TBS. The drug irenotecan (CPT-11) (LKT Laboratories) was injected IP (100 mg/kg) once a week for four weeks from the initiation of antibody treatment. Control animals received a class matched irrelevant human IgG antibody. Tumor measurements were performed at regular intervals using Vernier calipers, measuring height, width, and length and calculated to determine the total tumor volume. The study was terminated when control tumors reached 3000 mm . As shown in Fig. 23, doses of antibody 2F8 at either 0.5 mg or 1 mg every three days effected a significant inhibition (P < 0.05) of tumor growth in this model. There was no statistical difference between the tumor sizes from groups treated with 0.5 or 1 mg 2F8 and the responses were similar to treatment with CPT-11 alone. When 2F8 and CPT-11 were given together the combination resulted in greater inhibition of tumor growth (72% decrease), demonstrating that anti-IGF-IR therapy could enhance the anti-tumor activity of the chemotherapeutic agent CPT-11 on tumor growth.
Anti-tumor activity of antibody A12 on human colorectal tumors in vivo. [0146] Antibody A12 possesses a 10-fold higher affinity for the IGF-IR than antibody 2F8. Since significant tumor inhibition was observed in vivo with antibody 2F8, we investigated the activity of A12 on the growth of the human colorectal cancer line HT-29 in a mouse xenograft model. Tumors were induced as previously described, and antibody treatment initiated once tumors were established (200 mm3 size). Antibody treatment was then given at a concentration of 1 mg, 100 μg, or 10 μg every three days throughout the duration of the experiment. Ten animals were used per treatment group and control animals received a class match IgG control antibody. As shown in Fig. 24, antibody A12 effected a 74% reduction in tumor growth compared to control (P < 0.05). This demonstrated that A12 was effective as a single therapeutic at inhibiting colorectal tumor growth in this xenograft model. A clear dose-response effect was noted in this experiment. Anti-tumor activity was also observed with a dose of 100 μg A12.
Activity of antibody A12 on human breast cancer in vivo in a xenosraft tumor model. [0147] Antibody A12 exhibited strong inhibitory activity on the IGF-dependent mitogenic stimulation and proliferation of MCF7 cells in vitro. In order to assess its activity on MCF7 tumor growth in vivo, a mouse xenograft tumor model was utilized. MCF7 cells were originally isolated from an estrogen-dependent human tumor and require exogenously added estrogen for maintenance and growth in vivo. Nude mice were implanted with biodegradable estrogen pellets (0.72mg 17-β -estradiol/pellet, 60 day release). In addition, at the time of subcutaneous tumor cell injection, the mice were also injected in the right flank subcutaneously with 0.5 mg of estradiol in a 50 μl suspension of sesame seed oil. Tumors were allowed to establish a size of approximately 150 mm before antibody treatment was initiated. Antibody was injected at 1 mg, 100 μg, and 10 μg doses every three days and continued for the duration of the experiment. At 29 days, treatment of animals with 1 mg of A12 effected an 89% reduction in tumor growth (Fig. 25). Minimal growth was apparent for the established tumors in this treatment group. A dose dependent response was noted for A12 treatments in this model. The study demonstrated that A12 was effective in significantly reducing the growth of a human breast cancer cell line in vivo. Further, treating with antibody alone at 1 mg/dose, tumor regression was observed and continued to the termination of the study (50 days).
Efficacy ofA12 in combination with CPT-11 or gemcitabine in BxPC-3 pancreatic carcinoma xenografts [0148] Athymic nu/nu mice were injected subcutaneously with 2 x 10s BxPC-3 human pancreatic carcinoma cells mixed 1 : 1 with Matrigel. Twenty days later, when tumors reached 200-300mm3, mice were randomized and divided into treatment groups: 1) TBS control; 2) mAb A12 at lmg/dose, 3 times per week; 3) 1 mg irinotecan, once every 7 days; 4) mAb A12 + irinotecan; 5) 2.5 mg gemcitabine, once every 7 days; 6) mAb A12 + gemcitabine. All treatments were administered by intraperitoneal injection. Tumor measurements were recorded twice weekly using the formula Volume = (ττ/6) Ix w . [0149] At day 18 three animals per group were sacrificed and the tumors resected for midpoint histological evaluation. Treatment and tumor measurements continued on the remaining animals until day 44 with the exception of the TBS group. This control group was sacrificed at day 37 due to tumor ulceration and necrosis. At termination of the study final tumor measurements were recorded, animals sacrificed, and four per group had tumors resected for endpoint histological evaluation. [0150] BxPC-3 tumors were very responsive to mAb A12, and 2 of eight animals had partial tumor regressions after five weeks of treatment. The response of BxPC-3 tumors to irinotecan or gemcitabine alone was comparable to the antibody, but there were no tumor regressions. Antibody A12, when combined with irinotecan or gemcitabine was more effective than any agent alone (Fig. 26), with irinotecan + Mab A12 being the more effective combination. For combined A12 and irinotecan, three of nine animals had partial tumor regressions. However, there were no tumor regressions among the animals given irinotecan. (Table 5).
Figure imgf000044_0001
'Regressions defined as individual tumor volumes < day 1 of treatment.
Efficacy ofA12 in combination with CPT-11 or paclitaxel in HT-29 colorectal cancer xenografts [0151] Female thymic nu/nu mice were injected subcutaneously with HT-29 human colon carcinoma cell suspension, at 5 x 10 cells in 0.4 ml mixed 1:1 with Matrigel. When tumors reached ~200mm3 mice were randomized and divided into treatment groups: 1) TBS control; 2) mAb A12 at lmg/dose, 3 times per week; 3) 2 mg irinotecan, once every 7 days; 4) mAb A12 + irinotecan; 5) 121 μg paclitaxel in 0.2 ml, once evrey 7 days; 6) mAb A12 + paclitaxel. All treatments were administered by intraperitoneal injection. [0152] Treatment continued for six weeks and tumor measurements were recorded twice weekly using the formula Volume = (n 6) I x w2. At day 21 four animals per group were sacrificed and the tumors resected for histological evaluation. Treatment and tumor measurements continued on the remaining animals until day 40. At that time final tumor measurements were recorded, animals sacrificed, and four per group had tumor resected for endpoint histological evaluation. [0153] Single-agent mAb A12, CPT-11, or paclitaxel significantly (E < 0.02) inhibited the growth of HT-29 xenografts compared to the TBS control group (Fig. 27). Combination therapy of mAb A12 with either CPT-11 or paclitaxel showed a significant inhibition (E < 0.003) in tumor growth compared to either treatment alone. [0154] No tumor regressions were observed in the single-therapy groups. Control tumors in situ appeared highly vascularized at day 22 as did single-therapy groups at day 40. However, combination therapy with CPT-11 + mAb A12 resulted in partial tumor regressions in six out of eight animals, and combination therapy with paclitaxel and mAb A12 resulted in regressions in three of eight animals.
Table 6. Inhibition of Growth of HT-29 Xenografts mAb A12 CPT-11 paclitaxel A12 + CPT-11 A12 + paclitaxel Day T/C % T/C % T/C % T/C % Regressions* T/C % Regressions 12 69 63 66 52 0 / 12 48 1 /12 15 62 59 59 42 0 / 12 40 3 /12 19 64 61 62 30 3 / 12 32 5 /12 22 58 52 56 24 4 / 8 28 4 / 8 26 62 47 54 17 6 / 8 27 3 / 8 29 61 45 55 14 6 / 8 24 3 / 8 33 58 40 52 11 5 / 8 22 3 / 8 36 53 40 45 9 5 / 8 22 2 / 8 40 54 41 52 10 2 / 6 26 2 / 8 T/C % is tumor growth inhibition relative to control 'Regressions defined as individual tumor volumes < day 1 of treatment.
[0155] It is understood and expected that variations in the principles of invention herein disclosed may be made by one skilled in the art and it is intended that such modifications are to be included within the scope of the present invention.

Claims

What is claimed is:
1. An isolated human antibody or fragment thereof that specifically binds to insulin-like growth factor-I receptor (IGF-IR) and has at least one property selected from the group consisting of (i) inhibits binding of IGF-I or IGF-II to IGF-IR; (ii) neutralizes activation of IGF-IR by IGF-I or IGF-II; (iii) reduces IGF-IR surface receptor by at least about 80%; and (iv) binds to IGF-IR with a Kd of about 3 x 10"10 M"1 or less.
2. The antibody or antibody fragment of Claim 1 , which has all of said properties.
3. The antibody or antibody fragment of Claim 1 , wherein the antibody or antigen binding fragment thereof reduces surface IGF-IR by at least about 85%.
4. The antibody or antibody fragment of Claim 1, wherein the antibody or antigen binding fragment thereof reduces surface IGF-IR by at least about 90%.
5. The antibody or antibody fragment of Claim 1, which binds to IGF-IR with a K of about 1 x 10"10 M"1 or less.
6. The antibody or antibody fragment of Claim 1 , which binds to IGF-IR with a K of about 5 x 10 M"1 or less.
7. The antibody or antibody fragment of Claim 1, which inhibits phosphorylation of a downstream substrate of IFG-IR.
8. The antibody or antibody fragment of Claim 7, wherein the downstream substrate is selected from the group consisting of MAPK, Akt, and IRS-2, and phosphorylation is inhibited by about 50% or more.
9. The antibody or antibody fragment of Claim 1 , which promotes tumor regression in vivo.
10. The antibody or antibody fragment of Claim 1 , which promotes tumor regression in vivo when administered with an anti-neoplastic agent.
11. The antibody or antibody fragment of Claim 1 , which competes for binding to IGF-IR with an antibody selected from the group consisting of the antibody having a heavy chain variable domain represented by SEQ ID NO:2 and a light chain variable domain represented by SEQ ID NO:6; and the antibody having a heavy chain variable domain represented by SEQ ID NO:2 and a light chain variable domain represented by SEQ ID NO:10.
12. The antibody or antibody fragment of Claim 1 , which specifically binds to insulin-like growth factor-I receptor (IGF-IR) and comprises at least one complementarity- determining region (CDR) having an amino acid sequence selected from SEQ ID NO: 13 at VHCDRI, SEQ ID NO: 15 at VHCDR2, SEQ ID NO: 17 at VHCDR3, SEQ ID NO 19 at VLCDRI, SEQ ID NO:21 at VLCDR2, SEQ ID NO:23 at VLCDR3, SEQ ID NO 25 at VLCDRI, SEQ ID NO:27 at VLCDR2, and SEQ ID NO:29 at VLCDR3.
13. The antibody or antigen binding fragment of Claim 1 , which comprises SEQ ID NO:13 at VHCDR1, SEQ ID NO:15 at VHCDR2, and SEQ ID NO:17 at VHCDR3.
14. The antibody or antigen binding fragment of Claim 1 , which comprises SEQ ID NO 19 at VLCDR1, SEQ ID NO:21 at VLCDR2, and SEQ ID NO:23 at VLCDR3.
15. The antibody or antigen binding fragment of Claim 1 , which comprises SEQ ID NO 25 at V CDR1, SEQ ID NO:27 at VLCDR2. and SEQ ID NO:29 at VLCDR3.
16. The antibody of Claim 1, wherein the heavy chain variable domain has at least 90% sequence homology to SEQ ID NO:2.
17. The antibody of Claim 1, wherein the light chain variable domain has at least 90% sequence homology to SEQ ID NO:6.
18. The antibody of Claim 1, wherein the light chain variable domain has at least 90% sequence homology to SEQ ID NO: 10.
19. An isolated nucleic acid encoding a polypeptide selected from the group consisting of:
SEQ ID NO:2 from about amino acid residue 1 to about amino acid residue 130; SEQ ID NO:6 from about amino acid residue 1 to about amino acid residue 109; and SEQ ID NO: 10 from about amino acid residue 1 to about amino acid residue 109.
20. The isolated nucleic acid of Claim 19, selected from the group consisting of: SEQ ID NO:l from about nucleotide 1 to about nucleotide 390;
SEQ ID NO:5 from about nucleotide 1 to about nucleotide 327; and SEQ ID NO:9 from about nucleotide 1 to about nucleotide 327.
21. A recombinant vector comprising a nucleic acid of Claim 19.
22. A host cell comprising the vector of Claim 21.
23. A pharmaceutical composition comprising the antibody or antibody fragment of any one of Claims 1 to 18 and a pharmaceutically acceptable carrier.
24. A conjugate comprising the antibody or antibody fragment of any one of Claims 1 to 18 linked to a cytotoxic agent.
25. A conjugate comprising the antibody or antibody fragment of any one of Claims 1 to 18 linked to a label.
26. A therapeutic composition effective to inhibit growth of human tumor cells that express IGF-IR, which composition comprises the antibody or antigen binding fragment of any one of Claims 1 to 18.
27. The therapeutic composition of Claim 26, which further comprises an anti- neoplastic agent.
28. The therapeutic composition of Claim 27, wherein the anti-neoplastic agent is an inhibitor of topoisomerase I or topoisomerase II.
29. The therapeutic composition of Claim 27, wherein the anti-neoplastic agent is selected from the group consisting of irinotecan, camptothecan, and etoposide.
30. A therapeutic composition effective to promote regression of human tumors that express IGF-IR, which composition comprises the antibody or antibody fragment of any one of Claims 1 to 18.
31. The therapeutic composition of Claim 30, which further comprises an anti- neoplastic agent.
32. The therapeutic composition of Claim 31 , wherein the anti-neoplastic agent is an inhibitor of topoisomerase I or topoisomerase II.
33. The therapeutic composition of Claim 31 , wherein the anti-neoplastic agent is selected from the group consisting of irinotecan, camptothecan, or etoposide.
34. A method of neutralizing the activation of IGF-IR, which comprises administering to a mammal an effective amount of the antibody or antibody fragment of any one of Claims 1 to 18.
35. A method of treating a proliferative disorder comprising the step of administering an effective amount of the antibody or antibody fragment of any one of Claims 1 to 18.
36. The method of Claim 35, wherein the proliferative disorder is selected from the group consisting of acromegaly, retinal neovascularization, and psoriasis.
37. A method of inhibiting the growth of a cell that expresses IGF-IR, which comprises contacting the cell with an effective amount of the antibody or antibody fragment of any one of Claims 1 to 18.
38. The method of Claim 35, which further comprises contacting the cell with an effective amount of an anti-neoplastic agent.
39. The method of Claim 38, wherein the anti-neoplastic agent is an inhibitor of topoisomerase I or topoisomerase II.
40. The method of Claim 38, wherein the anti-neoplastic agent is selected from the group consisting of irinotecan, camptothecan, and etoposide.
41. A method of reducing tumor growth which comprises administering to a mammal an effective amount of the antibody or antibody fragment of any one of Claims 1 to 18.
42. The method of Claim 41, which further comprises administering an effective amount of an anti-neoplastic agent.
43. The method of Claim 42, wherein the anti-neoplastic agent is an inhibitor of topoisomerase I or topoisomerase II.
44. The method of Claim 42, wherein the anti-neoplastic agent is selected from the group consisting of irinotecan, camptothecan, and etoposide.
45. A method of promoting tumor regression which comprises administering to a mammal an effective amount of the antibody or antibody fragment of any one of Claims 1 to 18.
46. The method of Claim 45, which further comprises administering an effective amount of an anti-neoplastic agent.
47. The method of Claim 46, wherein the anti-neoplastic agent is an inhibitor of topoisomerase I or topoisomerase II.
48. The method of Claim 46, wherein the anti-neoplastic agent is selected from the group consisting of irinotecan, camptothecan, and etoposide.
49. The method of any one of Claims 41 to 48, wherein the tumor is a breast tumor, colorectal tumor, pancreatic tumor, ovarian tumor, lung tumor, prostate tumor, bone or soft tissue sarcoma or myeloma.
50. A method of inhibiting the growth of a cell that expresses IGF-IR, which comprises contacting the cell with an effective amount of an agent that is an inhibitor of topoisomerase I or topoisomerase II and an antibody or antigen binding fragment thereof that specifically binds to IGF-IR and has at least one property selected from the group consisting of (i) inhibits binding of IGF-I or IGF-II to IGF-IR; (ii) neutralizes activation of IGF-IR by IGF-I or IGF-II; (iii) reduces IGF-IR surface receptor; and (iv) binds to IGF-IR with a Kd of about 1 x 10"10 M"1 or less.
51. A method of reducing growth of a tumor that expresses IGF-IR, which comprises contacting the cell with an effective amount of an agent that is an inhibitor of topoisomerase I or topoisomerase II and an antibody or antigen binding fragment thereof that specifically binds to IGF-IR and has at least one property selected from the group consisting of (i) inhibits binding of IGF-I or IGF-II to IGF-IR; (ii) neutralizes activation of IGF-IR by IGF-I or IGF-II; (iii) reduces IGF-IR surface receptor by at least about 80%; and (iv) binds to IGF-IR with a K of about 1 x 10"10 M"1 or less.
52. A method of promoting regression of a tumor that expresses IGF-IR, which comprises contacting the cell with an effective amount of an agent that is an inhibitor of topoisomerase I or topoisomerase II and an antibody or antigen binding fragment thereof that specifically binds to IGF-IR and has at least one property selected from the group consisting of (i) inhibits binding of IGF-I or IGF-II to IGF-IR; (ii) neutralizes activation of IGF-IR by IGF-I or IGF-II; (iii) reduces IGF-IR surface receptor by at least about 80%; and (iv) binds to IGF-IR with a Kd of about 1 x 10'10 M"1 or less.
53. The method of any one of Claims 50 to 52, wherein the agent is selected from the group consisting of irinotecan, camptothecan, and etoposide.
54. The method of any one of Claims 50 to 52, wherein the antibody or antibody fragment is human.
55. The method of any one of Claims 50 to 52, wherein the antibody or antibody fragment is humanized.
56. The method of any one of Claims 51 and 52, wherein the tumor is a breast tumor, colorectal tumor, pancreatic tumor, ovarian tumor, lung tumor, prostate tumor, bone or soft tissue sarcoma or myeloma.
PCT/US2004/013852 2003-05-01 2004-05-03 Fully human antibodies directed against the human insulin-like growth factor-1 receptor WO2005016970A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CA2524305A CA2524305C (en) 2003-05-01 2004-05-03 Fully human antibodies directed against the human insulin-like growth factor-1 receptor
EP04775941.0A EP1622942B1 (en) 2003-05-01 2004-05-03 Fully human antibodies directed against the human insulin-like growth factor-1 receptor
JP2006514278A JP2007535895A (en) 2003-05-01 2004-05-03 Fully human antibody against human insulin-like growth factor-1 receptor
ES04775941.0T ES2527871T3 (en) 2003-05-01 2004-05-03 Fully human antibodies directed against human insulin-like growth factor 1 receptor
US10/555,407 US7638605B2 (en) 2003-05-01 2004-05-03 Fully human antibodies directed against the human insulin-like growth factor-1 receptor
US12/640,671 US7968093B2 (en) 2003-05-01 2009-12-17 Fully human antibodies directed against the human insulin-like growth factor-1 receptor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US46717703P 2003-05-01 2003-05-01
US60/467,177 2003-05-01

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/555,407 A-371-Of-International US7638605B2 (en) 2003-05-01 2004-05-03 Fully human antibodies directed against the human insulin-like growth factor-1 receptor
US12/640,671 Continuation US7968093B2 (en) 2003-05-01 2009-12-17 Fully human antibodies directed against the human insulin-like growth factor-1 receptor

Publications (2)

Publication Number Publication Date
WO2005016970A2 true WO2005016970A2 (en) 2005-02-24
WO2005016970A3 WO2005016970A3 (en) 2005-04-21

Family

ID=34193001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/013852 WO2005016970A2 (en) 2003-05-01 2004-05-03 Fully human antibodies directed against the human insulin-like growth factor-1 receptor

Country Status (6)

Country Link
US (2) US7638605B2 (en)
EP (2) EP2365001A3 (en)
JP (2) JP2007535895A (en)
CA (1) CA2524305C (en)
ES (1) ES2527871T3 (en)
WO (1) WO2005016970A2 (en)

Cited By (143)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006008639A1 (en) * 2004-07-16 2006-01-26 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-igf-1r antibody
WO2006060419A3 (en) * 2004-12-03 2006-08-24 Schering Corp Biomarkers for pre-selection of patients for anti-igf1r therapy
FR2888850A1 (en) * 2005-07-22 2007-01-26 Pierre Fabre Medicament Sa New isolated antibody that binds human insulin-like growth factor receptor, useful for treatment, prevention and diagnosis of e.g. cancer, psoriasis and arteriosclerosis
WO2007042289A2 (en) * 2005-10-11 2007-04-19 Ablynx N.V. Nanobodies™ and polypeptides against egfr and igf-ir
WO2007092453A2 (en) 2006-02-03 2007-08-16 Imclone Systems Incorporated Igf-ir antagonists as adjuvants for treatment of prostate cancer
WO2007070432A3 (en) * 2005-12-13 2007-11-29 Amgen Fremont Inc Binding proteins specific for insulin-like growth factors and uses thereof
EP1909819A2 (en) * 2005-06-17 2008-04-16 Imclone Systems, Inc. Receptor antagonists for treatment of metastatic bone cancer
WO2008076278A2 (en) 2006-12-13 2008-06-26 Schering Corporation Methods of cancer treatment with igf1r inhibitors
US20080193445A1 (en) * 2002-01-18 2008-08-14 Liliane Goetsch Novel anti-IGF-IR antibodies and uses thereof
WO2008098917A2 (en) * 2007-02-14 2008-08-21 Glaxo Group Limited Novel antibodies against igf-ir
WO2008144345A2 (en) 2007-05-17 2008-11-27 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
EP1999149A2 (en) * 2006-03-28 2008-12-10 Biogen Idec MA Inc. Anti-igf-ir antibodies and uses thereof
WO2008152422A2 (en) * 2007-06-12 2008-12-18 Astrazeneca Ab Binding proteins specific for insulin-like growth factors and uses thereof-909
WO2008116103A3 (en) * 2007-03-22 2009-01-08 Imclone Systems Inc Stable antibody formulations
WO2009032782A2 (en) * 2007-08-28 2009-03-12 Biogen Idec Ma Inc. Compositions that bind multiple epitopes of igf-1r
WO2009080278A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Stability testing of antibodies
EP2104514A2 (en) * 2006-11-29 2009-09-30 Imclone LLC Insulin-like growth factor-1 receptor antagonists for modulation of weight and liposity
US7667021B2 (en) 2002-05-24 2010-02-23 Schering Corporation Neutralizing human anti-IGFR antibody
WO2009137378A3 (en) * 2008-05-05 2010-03-04 Schering Corporation Sequential administration of chemotherapeutic agents for treatment of cancer
JP2010518140A (en) * 2007-02-14 2010-05-27 グラクソ グループ リミテッド Novel antibody against IGF-1R
EP2236139A1 (en) 2009-03-31 2010-10-06 F. Hoffmann-La Roche AG Combination therapy of erlotinib with an anti-IGF-1R antibody, which does not inhibit binding of insulin to the insulin receptor
US7846724B2 (en) 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
US7871611B2 (en) 2004-12-22 2011-01-18 Amgen Inc. Compositions and methods relating to anti IGF-1 receptor antibodies
WO2011050333A1 (en) 2009-10-23 2011-04-28 Amgen Inc. Vial adapter and system
WO2011053779A2 (en) 2009-10-30 2011-05-05 Bristol-Myers Squibb Company Methods for treating cancer in patients having igf-1r inhibitor resistance
US7947811B2 (en) 2007-11-21 2011-05-24 Imclone Llc Antibodies that bind specifically to human RON protein
EP2332990A1 (en) 2004-03-19 2011-06-15 Imclone LLC Human anti-epidermal growth factor receptor antibody
WO2011083391A2 (en) 2010-01-05 2011-07-14 Pfizer Inc. Biomarkers for anti-igf-ir cancer therapy
WO2011101328A2 (en) 2010-02-18 2011-08-25 Roche Glycart Ag Treatment with a humanized igg class anti egfr antibody and an antibody against insulin like growth factor 1 receptor
US8017735B2 (en) 2003-11-21 2011-09-13 Schering Corporation Anti-IGFR1 antibody therapeutic combinations
US8062886B2 (en) 2003-11-12 2011-11-22 Schering Corporation Plasmid system for multigene expression
WO2011156373A1 (en) 2010-06-07 2011-12-15 Amgen Inc. Drug delivery device
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
EP2405270A1 (en) 2006-06-30 2012-01-11 Schering Corporation IGFBP2-Biomarker
US8178091B2 (en) 2007-05-21 2012-05-15 University Of Washington Compositions and methods for the treatment of respiratory disorders
WO2012106556A2 (en) 2011-02-02 2012-08-09 Amgen Inc. Methods and compositons relating to inhibition of igf-1r
WO2012135315A1 (en) 2011-03-31 2012-10-04 Amgen Inc. Vial adapter and system
US8318159B2 (en) 2008-12-12 2012-11-27 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US8383114B2 (en) 2007-09-27 2013-02-26 Amgen Inc. Pharmaceutical formulations
WO2013056069A1 (en) 2011-10-13 2013-04-18 Bristol-Myers Squibb Company Methods for selecting and treating cancer in patients with igf-1r/ir inhibitors
WO2013055873A1 (en) 2011-10-14 2013-04-18 Amgen Inc. Injector and method of assembly
WO2013071056A2 (en) 2011-11-11 2013-05-16 Duke University Combination drug therapy for the treatment of solid tumors
EP2631653A1 (en) 2012-02-24 2013-08-28 Charité - Universitätsmedizin Berlin Identification of modulators of binding properties of antibodies reactive with a member of the insulin receptor family
US8580254B2 (en) 2007-06-19 2013-11-12 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
WO2014015280A1 (en) 2012-07-20 2014-01-23 Novartis Pharma Ag Combination therapy of inhibitors for igf1 r and pi3k
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
EP2727941A1 (en) 2012-11-05 2014-05-07 MAB Discovery GmbH Method for the production of multispecific antibodies
EP2727936A1 (en) * 2006-11-22 2014-05-07 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
WO2014067642A1 (en) 2012-11-05 2014-05-08 Mab Discovery Gmbh Method for the production of multispecific antibodies
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2014081780A1 (en) 2012-11-21 2014-05-30 Amgen Inc. Drug delivery device
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
WO2014144096A1 (en) 2013-03-15 2014-09-18 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
WO2014149357A1 (en) 2013-03-22 2014-09-25 Amgen Inc. Injector and method of assembly
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
WO2015061386A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Injector and method of assembly
WO2015061389A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Drug delivery system with temperature-sensitive control
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
WO2015171777A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
WO2015187797A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Controllable drug delivery system and method of use
US9234028B2 (en) 2008-02-14 2016-01-12 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
WO2016061220A2 (en) 2014-10-14 2016-04-21 Amgen Inc. Drug injection device with visual and audio indicators
WO2016100781A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with proximity sensor
WO2016100055A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with live button or user interface field
CN105884900A (en) * 2011-04-19 2016-08-24 梅里麦克制药股份有限公司 Monospecific and bispecific anti-IGF-1R and anti-ERBB3 antibodies
WO2016133947A1 (en) 2015-02-17 2016-08-25 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
WO2016138434A1 (en) 2015-02-27 2016-09-01 Amgen Inc. Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
US9562089B2 (en) 2010-05-26 2017-02-07 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
WO2017100501A1 (en) 2015-12-09 2017-06-15 Amgen Inc. Auto-injector with signaling cap
WO2017120178A1 (en) 2016-01-06 2017-07-13 Amgen Inc. Auto-injector with signaling electronics
WO2017160799A1 (en) 2016-03-15 2017-09-21 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
WO2017189089A1 (en) 2016-04-29 2017-11-02 Amgen Inc. Drug delivery device with messaging label
WO2017192287A1 (en) 2016-05-02 2017-11-09 Amgen Inc. Syringe adapter and guide for filling an on-body injector
WO2017197222A1 (en) 2016-05-13 2017-11-16 Amgen Inc. Vial sleeve assembly
WO2017200989A1 (en) 2016-05-16 2017-11-23 Amgen Inc. Data encryption in medical devices with limited computational capability
WO2017209899A1 (en) 2016-06-03 2017-12-07 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
WO2018004842A1 (en) 2016-07-01 2018-01-04 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
WO2018081234A1 (en) 2016-10-25 2018-05-03 Amgen Inc. On-body injector
WO2018136398A1 (en) 2017-01-17 2018-07-26 Amgen Inc. Injection devices and related methods of use and assembly
WO2018151890A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Drug delivery device with sterile fluid flowpath and related method of assembly
WO2018152073A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Insertion mechanism for drug delivery device
WO2018165143A1 (en) 2017-03-06 2018-09-13 Amgen Inc. Drug delivery device with activation prevention feature
WO2018165499A1 (en) 2017-03-09 2018-09-13 Amgen Inc. Insertion mechanism for drug delivery device
WO2018164829A1 (en) 2017-03-07 2018-09-13 Amgen Inc. Needle insertion by overpressure
EP3381445A2 (en) 2007-11-15 2018-10-03 Amgen Inc. Aqueous formulation of antibody stablised by antioxidants for parenteral administration
WO2018183039A1 (en) 2017-03-28 2018-10-04 Amgen Inc. Plunger rod and syringe assembly system and method
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2018226515A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
WO2018226565A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Torque driven drug delivery device
WO2018236619A1 (en) 2017-06-22 2018-12-27 Amgen Inc. Device activation impact/shock reduction
WO2018237225A1 (en) 2017-06-23 2018-12-27 Amgen Inc. Electronic drug delivery device comprising a cap activated by a switch assembly
WO2019014014A1 (en) 2017-07-14 2019-01-17 Amgen Inc. Needle insertion-retraction system having dual torsion spring system
WO2019018169A1 (en) 2017-07-21 2019-01-24 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
WO2019022950A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with container access system and related method of assembly
WO2019022951A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with gear module and related method of assembly
WO2019032482A2 (en) 2017-08-09 2019-02-14 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
WO2019036181A1 (en) 2017-08-18 2019-02-21 Amgen Inc. Wearable injector with sterile adhesive patch
WO2019040548A1 (en) 2017-08-22 2019-02-28 Amgen Inc. Needle insertion mechanism for drug delivery device
WO2019070552A1 (en) 2017-10-06 2019-04-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
WO2019070472A1 (en) 2017-10-04 2019-04-11 Amgen Inc. Flow adapter for drug delivery device
WO2019074579A1 (en) 2017-10-09 2019-04-18 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
WO2019090086A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Systems and approaches for sterilizing a drug delivery device
WO2019089178A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Drug delivery device with placement and flow sensing
WO2019090303A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Fill-finish assemblies and related methods
WO2019094138A1 (en) 2017-11-10 2019-05-16 Amgen Inc. Plungers for drug delivery devices
WO2019099322A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Autoinjector with stall and end point detection
WO2019099324A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Door latch mechanism for drug delivery device
EP3498323A2 (en) 2011-04-20 2019-06-19 Amgen Inc. Autoinjector apparatus
US10377828B2 (en) 2013-03-07 2019-08-13 Boehringer Ingelheim International Gmbh Combination therapy for neoplasia treatment
WO2019231582A1 (en) 2018-05-30 2019-12-05 Amgen Inc. Thermal spring release mechanism for a drug delivery device
WO2019231618A1 (en) 2018-06-01 2019-12-05 Amgen Inc. Modular fluid path assemblies for drug delivery devices
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
WO2020023444A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020023451A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020028009A1 (en) 2018-07-31 2020-02-06 Amgen Inc. Fluid path assembly for a drug delivery device
WO2020068476A1 (en) 2018-09-28 2020-04-02 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
WO2020068623A1 (en) 2018-09-24 2020-04-02 Amgen Inc. Interventional dosing systems and methods
WO2020072846A1 (en) 2018-10-05 2020-04-09 Amgen Inc. Drug delivery device having dose indicator
WO2020072577A1 (en) 2018-10-02 2020-04-09 Amgen Inc. Injection systems for drug delivery with internal force transmission
WO2020081479A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Drug delivery device having damping mechanism
WO2020081480A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Platform assembly process for drug delivery device
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020092056A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial needle retraction
WO2020091956A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020219482A1 (en) 2019-04-24 2020-10-29 Amgen Inc. Syringe sterilization verification assemblies and methods
WO2021041067A2 (en) 2019-08-23 2021-03-04 Amgen Inc. Drug delivery device with configurable needle shield engagement components and related methods
US11021728B2 (en) 2009-10-26 2021-06-01 Hoffmann-La Roche Inc. Method for the production of a glycosylated immunoglobulin
WO2022246055A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container
WO2023138620A1 (en) * 2022-01-19 2023-07-27 黄浩旻 Anti-pdl1/igf1r bispecific antibody, preparation method therefor and use thereof

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20020801A1 (en) 2001-01-05 2002-09-06 Pfizer ANTIBODIES AGAINST INSULIN-LIKE GROWTH FACTOR RECEPTOR
US7559282B2 (en) * 2002-10-16 2009-07-14 Robert Austin Monorail sortation system
US7579157B2 (en) * 2003-07-10 2009-08-25 Hoffmann-La Roche Inc. Antibody selection method against IGF-IR
WO2005072292A2 (en) * 2004-01-24 2005-08-11 Hugh Mctavish Methods for enhancing radiation therapy
NZ565654A (en) * 2005-07-18 2010-10-29 Bipar Sciences Inc Use of iodonitrobenzamide compounds for the treatment of ovarian cancer
US20100279327A1 (en) * 2006-06-12 2010-11-04 Bipar Sciences, Inc. Method of treating diseases with parp inhibitors
WO2008030883A2 (en) * 2006-09-05 2008-03-13 Bipar Sciences, Inc. Treatment of cancer
CA2662337A1 (en) * 2006-09-05 2008-03-13 Bipar Sciences, Inc. Inhibition of fatty acid synthesis by parp inhibitors and methods of treatment thereof
WO2009032145A1 (en) * 2007-08-28 2009-03-12 Biogen Idec Ma Inc. Anti-igf-1r antibodies and uses thereof
SG185952A1 (en) * 2007-11-12 2012-12-28 Bipar Sciences Inc Treatment of breast cancer with 4-iodo-3-nitrobenzamide in combination with anti-tumor agents
WO2009064444A2 (en) * 2007-11-12 2009-05-22 Bipar Sciences, Inc. Treatment of uterine cancer and ovarian cancer with a parp inhibitor alone or in combination with anti-tumor agents
US20090149397A1 (en) * 2007-12-07 2009-06-11 Bipar Sciences Treatment of cancer with combinations of topoisomerase inhibitors and parp inhibitors
US20090291088A1 (en) * 2008-04-11 2009-11-26 Biogen Idec Ma Inc. Therapeutic combinations of anti-igf-1r antibodies and other compounds
TW201032796A (en) * 2009-02-04 2010-09-16 Bipar Sciences Inc Treatment of lung cancer with a PARP inhibitor in combination with a growth factor inhibitor
CN102481361B (en) 2009-04-16 2014-10-22 默沙东公司 Compositions and methods for treating cancer
US9056907B2 (en) 2009-10-07 2015-06-16 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human domain antibodies against components of the human insulin-like growth factor (IGF) system
WO2011139973A2 (en) * 2010-05-03 2011-11-10 Abbott Laboratories Methods of inhibiting fibrosis using anti-pai-1 antibodies
KR101758152B1 (en) * 2010-10-22 2017-07-14 삼성전자주식회사 Apparatus and method for handling network loss in wireless access system
WO2013060872A1 (en) * 2011-10-27 2013-05-02 Boehringer Ingelheim International Gmbh Anticancer combination therapy
US9629801B2 (en) * 2014-01-10 2017-04-25 Wisconsin Alumni Research Foundation Blood-brain barrier targeting antibodies
US20210332141A1 (en) 2017-08-30 2021-10-28 Amgen Inc. Insulin-like growth factor-1 receptor (igf-1r) binding proteins and methods of use
US11279698B2 (en) 2018-11-20 2022-03-22 Cornell University Macrocyclic complexes of alpha-emitting radionuclides and their use in targeted radiotherapy of cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002053596A2 (en) 2001-01-05 2002-07-11 Pfizer Inc. Antibodies to insulin-like growth factor i receptor

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5585344A (en) 1984-03-19 1996-12-17 The Rockefeller University Liver-derived receptors for advanced glycosylation endproducts and uses thereof
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4774321A (en) 1986-12-01 1988-09-27 Massachusetts Institute Of Technology DP100 EGF and insulin-binding protein from Drosophila cells
ATE90690T1 (en) 1987-04-06 1993-07-15 Celtrix Pharma HUMAN SOMATOMEDIN CARRIER PROTEIN SUBUNITS AND METHODS FOR THEIR PRODUCTION.
US5200509A (en) 1987-04-06 1993-04-06 Celtrix Pharmaceuticals, Inc. Human somatomedin carrier protein subunits and process for producing them; recombinant DNA molecules, hosts, processes and human somatomedin carrier protein-like polypeptides
ATE243754T1 (en) 1987-05-21 2003-07-15 Micromet Ag MULTIFUNCTIONAL PROTEINS WITH PREDEFINED TARGET
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
IL89489A0 (en) 1988-03-09 1989-09-10 Hybritech Inc Chimeric antibodies directed against human carcinoembryonic antigen
JPH02503867A (en) 1988-04-15 1990-11-15 プロテイン デザイン ラブズ インコーポレーテッド IL-2 receptor-specific chimeric antibody
ATE119198T1 (en) 1988-04-16 1995-03-15 Celltech Ltd METHOD FOR PRODUCING PROTEINS USING RECOMBINANT DNA.
AU4128089A (en) 1988-09-15 1990-03-22 Rorer International (Overseas) Inc. Monoclonal antibodies specific to human epidermal growth factor receptor and therapeutic methods employing same
GB8826451D0 (en) 1988-11-11 1988-12-14 Sandoz Ltd Improvements in/relating to organic compounds
IL92816A0 (en) 1988-12-22 1990-09-17 Biogrowth Inc Recombinant dna molecules,hosts,processes and human somatomedin carrier protein-like polypeptides
US5571714A (en) 1988-12-22 1996-11-05 Celtrix Pharmaceuticals, Inc. Monoclonal antibodies which bind both transforming growth factors β1 and β2 and methods of use
US5705157A (en) 1989-07-27 1998-01-06 The Trustees Of The University Of Pennsylvania Methods of treating cancerous cells with anti-receptor antibodies
US5977307A (en) 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
JPH06503711A (en) 1990-08-28 1994-04-28 カイロン コーポレイション Novel insulin-like growth factor binding protein (IGFBP-4)
DK1024191T3 (en) 1991-12-02 2008-12-08 Medical Res Council Preparation of autoantibodies displayed on phage surfaces from antibody segment libraries
US5262308A (en) 1992-01-28 1993-11-16 Thomas Jefferson University Cell lines which constitutively express IGF-1 and IGF-1 R
AU4025193A (en) 1992-04-08 1993-11-18 Cetus Oncology Corporation Humanized C-erbB-2 specific antibodies
US5597563A (en) 1992-09-04 1997-01-28 Beschorner; William E. Method induction of antigen-specific immune tolerance
US5869337A (en) 1993-02-12 1999-02-09 President And Fellows Of Harvard College Regulated transcription of targeted genes and other biological events
EP0694069A1 (en) 1993-04-06 1996-01-31 Cedars-Sinai Medical Center Variant insulin-like growth factor i receptor subunits and methods for use thereof
WO1995019570A1 (en) 1994-01-14 1995-07-20 Genentech, Inc. Antagonists to insulin receptor tyrosine kinase inhibitor
US5532159A (en) 1994-04-01 1996-07-02 The Ohio State University Monoclonal antibody to canine placental oncofetal protein for detecting cancer
US5597700A (en) 1994-04-28 1997-01-28 California Research, Llc Method for detecting free insulin-like growth-factor-binding protein 1 and a test device for detecting the ruptures of fetal membranes using the above method
EP0764030B1 (en) 1994-06-24 2000-09-20 Vladimir P. Torchilin Composition containing autoantibodies for tumor therapy and prophylaxis
US5939269A (en) 1994-12-28 1999-08-17 The Regents Of The University Of California Antagonists to insulin receptor tyrosine kinase inhibitor
EP0822830B1 (en) * 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
US6084085A (en) 1995-11-14 2000-07-04 Thomas Jefferson University Inducing resistance to tumor growth with soluble IGF-1 receptor
AUPN999096A0 (en) 1996-05-22 1996-06-13 Northstar Biologicals Pty Ltd Peptides, antibodies, vaccines & uses thereof
US5798266A (en) 1996-08-27 1998-08-25 K-Quay Enterprises, Llc Methods and kits for obtaining and assaying mammary fluid samples for breast diseases, including cancer
US6071891A (en) 1996-11-22 2000-06-06 Regents Of The University Of Minnesota Insulin-like growth factor 1 receptors (IGF-1R) antisense oligonucleotide cells composition
ZA200007412B (en) 1998-05-15 2002-03-12 Imclone Systems Inc Treatment of human tumors with radiation and inhibitors of growth factor receptor tyrosine kinases.
SK17282000A3 (en) 1998-05-15 2002-04-04 Imclone Systems Incorporated Non-radiolabeled protein receptor tyrosine kinase inhibitor
US6875741B2 (en) 1998-09-02 2005-04-05 Renuka Pillutla Insulin and IGF-1 receptor agonists and antagonists
JP2002526419A (en) 1998-10-05 2002-08-20 ファーメクサ エイ/エス Novel methods for therapeutic vaccination
EP1006184A1 (en) 1998-12-03 2000-06-07 F. Hoffmann-La Roche Ag IGF-1 receptor interacting proteins (IIPs) genes coding therefor and uses thereof
US6316462B1 (en) 1999-04-09 2001-11-13 Schering Corporation Methods of inducing cancer cell death and tumor regression
US7329745B2 (en) 2000-06-13 2008-02-12 City Of Hope Single-chain antibodies against human insulin-like growth factor I receptor: expression, purification, and effect on tumor growth
US20030165502A1 (en) 2000-06-13 2003-09-04 City Of Hope Single-chain antibodies against human insulin-like growth factor I receptor: expression, purification, and effect on tumor growth
WO2001098493A1 (en) 2000-06-15 2001-12-27 Kyowa Hakko Kogyo Co., Ltd Insulin-like growth factor-binding protein
US8153121B2 (en) 2000-10-06 2012-04-10 Los Angeles Biomedical Research Institute at Harbor—UCLA Medical Center Diagnosis and therapy of antibody-mediated inflammatory autoimmune disorders
IL157705A0 (en) 2001-03-14 2004-03-28 Genentech Inc Igf antagonist peptides
US20040116330A1 (en) 2001-04-27 2004-06-17 Kenichiro Naito Preventive/therapeutic method for cancer
WO2004003019A2 (en) 2002-06-28 2004-01-08 Domantis Limited Immunoglobin single variant antigen-binding domains and dual-specific constructs
US7553485B2 (en) 2002-01-18 2009-06-30 Pierre Fabre Medicament Anti-IGF-IR and/or anti-insulin/IGF-I hybrid receptors antibodies and uses thereof
SI1461359T1 (en) 2002-01-18 2007-06-30 Pf Medicament Novel anti-igf-ir antibodies and uses thereof
US7241444B2 (en) 2002-01-18 2007-07-10 Pierre Fabre Medicament Anti-IGF-IR antibodies and uses thereof
ES2362931T3 (en) 2002-03-04 2011-07-15 Imclone Llc SPECIFIC HUMAN ANTIBODIES AGAINST KDR AND USES OF THE SAME.
IL165256A0 (en) * 2002-05-24 2005-12-18 Schering Corp Neutralizing human anti-igfr antibody
US8034904B2 (en) 2002-06-14 2011-10-11 Immunogen Inc. Anti-IGF-I receptor antibody
US7538195B2 (en) 2002-06-14 2009-05-26 Immunogen Inc. Anti-IGF-I receptor antibody
JP5138867B2 (en) 2002-08-01 2013-02-06 イミューノメディクス、インコーポレイテッド α-fetoprotein Immu31 antibody and fusion protein and method of use thereof
US20040102360A1 (en) 2002-10-30 2004-05-27 Barnett Stanley F. Combination therapy
NZ582210A (en) 2003-02-13 2011-04-29 Pfizer Prod Inc Uses of anti-insulin-like growth factor I receptor antibodies
CA2518980A1 (en) * 2003-03-14 2004-09-30 Pharmacia Corporation Antibodies to igf-i receptor for the treatment of cancers
ES2383014T3 (en) * 2003-04-02 2012-06-15 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor I and their uses
US7579157B2 (en) 2003-07-10 2009-08-25 Hoffmann-La Roche Inc. Antibody selection method against IGF-IR
EP1651663B1 (en) 2003-08-08 2017-05-17 Immunomedics, Inc. Bispecific antibodies for inducing apoptosis of tumor and diseased cells
OA13234A (en) 2003-08-13 2006-12-13 Pfizer Prod Inc Modified human IGF-1R antibodies.
JPWO2005027970A1 (en) 2003-09-24 2007-11-15 協和醗酵工業株式会社 Drugs for cancer treatment
TW200526684A (en) 2003-11-21 2005-08-16 Schering Corp Anti-IGFR1 antibody therapeutic combinations
ES2368741T3 (en) 2004-02-25 2011-11-21 Dana-Farber Cancer Institute, Inc. INHIBITORS OF THE RECEPTOR 1 OF THE INSULIN TYPE GROWTH FACTOR TO INHIBIT THE GROWTH OF TUMOR CELLS.
JP2008506681A (en) 2004-07-16 2008-03-06 ファイザー・プロダクツ・インク Combination therapy for non-blood malignant tumors using anti-IGF-1R antibody
FR2873699B1 (en) 2004-07-29 2009-08-21 Pierre Fabre Medicament Sa NOVEL ANTI-IGF ANTIBODIES IR RT USES THEREOF
WO2006060419A2 (en) 2004-12-03 2006-06-08 Schering Corporation Biomarkers for pre-selection of patients for anti-igf1r therapy
US20060134172A1 (en) 2004-12-21 2006-06-22 Alcon, Inc. Agents which regulate, inhibit, or modulate the activity and/or expression of lysyl oxidase (LOX) and LOX-like proteases as a unique means to both lower intraocular pressure and treat glaucomatous retinopathies/optic neuropathies
MY146381A (en) 2004-12-22 2012-08-15 Amgen Inc Compositions and methods relating relating to anti-igf-1 receptor antibodies
MX2007012817A (en) 2005-04-15 2007-12-12 Immunogen Inc Elimination of heterogeneous or mixed cell population in tumors.
WO2007000328A1 (en) 2005-06-27 2007-01-04 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Antibodies that bind to an epitope on insulin-like growth factor 1 receptor and uses thereof
US20070009970A1 (en) 2005-07-08 2007-01-11 Predicant Biosciences, Inc. Biological patterns for diagnosis and treatment of cancer
FR2888850B1 (en) 2005-07-22 2013-01-11 Pf Medicament NOVEL ANTI-IGF-IR ANTIBODIES AND THEIR APPLICATIONS
CN101291954B (en) 2005-08-26 2013-03-27 罗氏格黎卡特股份公司 Modified antigen binding molecules with altered cell signaling activity
EP1979001B1 (en) 2005-12-13 2012-04-11 Medimmune Limited Binding proteins specific for insulin-like growth factors and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002053596A2 (en) 2001-01-05 2002-07-11 Pfizer Inc. Antibodies to insulin-like growth factor i receptor

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
ARTEGA; OSBORNE, CANCER RES., vol. 49, 1989, pages 6237 - 41
D'AMBROSIO ET AL., CANCER RES., vol. 56, 1996, pages 4013 - 20
FURLANETTO ET AL., CANCER RES., vol. 53, 1993, pages 2522 - 26
HAYLOR ET AL., J. AM. SOC. NEPHROL., vol. 11, 2000, pages 2027 - 35
PARK; SMOLEN: "Advances in Protein Chemistry", 2001, ACADEMIC PRESS, pages: 360 - 421
PIETRZKOWSKI ET AL., CANCER RES., vol. 52, 1992, pages 6447 - 51
REISS ET AL., J. CELL. PHYS., vol. 181, 1999, pages 124 - 35
SCOTLANDI ET AL., CANCER RES., vol. 58, 1998, pages 4127 - 31
SCOTLANDI ET AL., INT. J. CANCER, vol. 101, 2002, pages 11 - 6
SEELY ET AL., BMC CANCER, vol. 2, 2002, pages 15

Cited By (256)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080193445A1 (en) * 2002-01-18 2008-08-14 Liliane Goetsch Novel anti-IGF-IR antibodies and uses thereof
US7847068B2 (en) 2002-05-24 2010-12-07 Schering Corporation Neutralizing human anti-IGFR antibody
US7667021B2 (en) 2002-05-24 2010-02-23 Schering Corporation Neutralizing human anti-IGFR antibody
US7851181B2 (en) 2002-05-24 2010-12-14 Schering Corporation Neutralizing human anti-IGFR antibody
US8062886B2 (en) 2003-11-12 2011-11-22 Schering Corporation Plasmid system for multigene expression
US8017735B2 (en) 2003-11-21 2011-09-13 Schering Corporation Anti-IGFR1 antibody therapeutic combinations
EP2332990A1 (en) 2004-03-19 2011-06-15 Imclone LLC Human anti-epidermal growth factor receptor antibody
WO2006008639A1 (en) * 2004-07-16 2006-01-26 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-igf-1r antibody
EP2322215A2 (en) 2004-07-16 2011-05-18 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-IGF-1R antibody
EP2322217A2 (en) 2004-07-16 2011-05-18 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-IGF-1R antibody
EP2335727A2 (en) 2004-07-16 2011-06-22 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-IGF-1R antibody
US7811562B2 (en) 2004-12-03 2010-10-12 Schering Corporation Biomarkers for pre-selection of patients for anti-IGF1R therapy
JP2008521907A (en) * 2004-12-03 2008-06-26 シェーリング コーポレイション Biomarkers for preselecting patients for anti-IGF1R treatment
WO2006060419A3 (en) * 2004-12-03 2006-08-24 Schering Corp Biomarkers for pre-selection of patients for anti-igf1r therapy
EP2281841A3 (en) * 2004-12-03 2013-10-23 Merck Sharp & Dohme Corp. Biomarkers for pre-selection of patients for anti-IGF1R therapy
EP2283831A3 (en) * 2004-12-03 2013-10-23 Merck Sharp & Dohme Corp. Biomakers for pre-selection of patients for anti-IGF1R therapy
US10035861B2 (en) 2004-12-22 2018-07-31 Amgen Inc. Compositions and methods relating to anti-IGF-1 receptor antibodies
US8460662B2 (en) 2004-12-22 2013-06-11 Amgen Inc. Compositions and methods relating to anti-IGF-1 receptor antibodies
EP2322551A2 (en) 2004-12-22 2011-05-18 Amgen, Inc Compositions comprising Anti-IGF-1R Antibodies and Methods for their Production
EP2322550A1 (en) 2004-12-22 2011-05-18 Amgen, Inc Compositions comprising anti-IGF-1R Antibodies and Methods for obtaining said Antibodies
US7871611B2 (en) 2004-12-22 2011-01-18 Amgen Inc. Compositions and methods relating to anti IGF-1 receptor antibodies
US8895008B2 (en) 2004-12-22 2014-11-25 Amgen Inc. Compositions and methods relating to anti-IGF-1 receptor antibodies
US8168409B2 (en) 2004-12-22 2012-05-01 Amgen Inc. Compositions and methods relating to anti-IGF-1 receptor antibodies
EP2100614A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
EP2505205A1 (en) 2005-06-17 2012-10-03 Imclone LLC Anti-PDGFR Alpha Antibodies
US8574578B2 (en) 2005-06-17 2013-11-05 Imclone Llc Antibodies against PDGFRα to inhibit tumor growth
EP2100618A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
EP1909819A2 (en) * 2005-06-17 2008-04-16 Imclone Systems, Inc. Receptor antagonists for treatment of metastatic bone cancer
JP2009501141A (en) * 2005-06-17 2009-01-15 イムクローン システムズ インコーポレイテッド Receptor antagonists for the treatment of metastatic bone cancer
EP1909819A4 (en) * 2005-06-17 2010-02-17 Imclone Llc Receptor antagonists for treatment of metastatic bone cancer
WO2007012614A3 (en) * 2005-07-22 2007-03-29 Pf Medicament Novel anti-igf-ir antibodies and uses thereof
US8361461B2 (en) 2005-07-22 2013-01-29 Pierre Fabre Medicament Anti-IGF-IR antibodies and uses thereof
AU2006274086B2 (en) * 2005-07-22 2012-09-13 Pierre Fabre Medicament Novel anti-IGF-IR antibodies and uses thereof
EP2380913A1 (en) * 2005-07-22 2011-10-26 Pierre Fabre Medicament Novel anti-IGF-IR antibodies and uses thereof
WO2007012614A2 (en) 2005-07-22 2007-02-01 Pierre Fabre Medicament Novel anti-igf-ir antibodies and uses thereof
FR2888850A1 (en) * 2005-07-22 2007-01-26 Pierre Fabre Medicament Sa New isolated antibody that binds human insulin-like growth factor receptor, useful for treatment, prevention and diagnosis of e.g. cancer, psoriasis and arteriosclerosis
WO2007042289A3 (en) * 2005-10-11 2007-10-04 Ablynx Nv Nanobodies™ and polypeptides against egfr and igf-ir
WO2007042289A2 (en) * 2005-10-11 2007-04-19 Ablynx N.V. Nanobodies™ and polypeptides against egfr and igf-ir
WO2007070432A3 (en) * 2005-12-13 2007-11-29 Amgen Fremont Inc Binding proteins specific for insulin-like growth factors and uses thereof
US7939637B2 (en) 2005-12-13 2011-05-10 Medimmune Limited Insulin-like growth factor antibodies and uses thereof
US8410058B2 (en) 2005-12-13 2013-04-02 Medimmune Limited Methods of using insulin-like growth factor antibodies to treat tumors
EP2404616A3 (en) * 2005-12-13 2012-02-29 AstraZeneca AB Binding proteins specific for insulin-like growth factors and uses thereof
AU2006326649B2 (en) * 2005-12-13 2011-03-24 Medimmune Limited Binding proteins specific for insulin-like growth factors and uses thereof
US9796779B2 (en) 2005-12-13 2017-10-24 Medimmune Limited Binding proteins specific for insulin-like growth factors and uses thereof
US9181336B2 (en) 2005-12-13 2015-11-10 Medimmune Limited Binding proteins specific for insulin-like growth factors and uses thereof
WO2007092453A2 (en) 2006-02-03 2007-08-16 Imclone Systems Incorporated Igf-ir antagonists as adjuvants for treatment of prostate cancer
CN101484587B (en) * 2006-02-03 2014-02-12 英克隆有限责任公司 IGF-IR antagonists as adjuvants for treatment of prostate cancer
US7972600B2 (en) 2006-02-03 2011-07-05 Imclone Llc IGF-IR antagonists as adjuvants for treatment of prostate cancer
JP2009532027A (en) * 2006-03-28 2009-09-10 バイオジェン・アイデック・エムエイ・インコーポレイテッド Anti-IGF-1R antibodies and uses thereof
US8147829B2 (en) 2006-03-28 2012-04-03 Biogen Idec Ma Inc. Anti-IGR-1R antibodies and uses thereof
EP1999149A4 (en) * 2006-03-28 2010-01-20 Biogen Idec Inc Anti-igf-ir antibodies and uses thereof
EP1999149A2 (en) * 2006-03-28 2008-12-10 Biogen Idec MA Inc. Anti-igf-ir antibodies and uses thereof
US8703919B2 (en) 2006-04-11 2014-04-22 Hoffmann-La Roche Inc. Glycosylated antibodies
US11673958B2 (en) 2006-04-11 2023-06-13 Hoffmann-La Roche Inc. Glycosylated antibodies
US7846724B2 (en) 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
EP2405270A1 (en) 2006-06-30 2012-01-11 Schering Corporation IGFBP2-Biomarker
EP3156415A1 (en) * 2006-11-22 2017-04-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including igf-ir
US11149077B2 (en) 2006-11-22 2021-10-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
US10221232B2 (en) 2006-11-22 2019-03-05 Bristol-Myers Squibb Company Methods of treating cancer by administering IGF-IR binding molecules
EP2727936A1 (en) * 2006-11-22 2014-05-07 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
EP2104514A4 (en) * 2006-11-29 2010-06-23 Imclone Llc Insulin-like growth factor-1 receptor antagonists for modulation of weight and liposity
EP2104514A2 (en) * 2006-11-29 2009-09-30 Imclone LLC Insulin-like growth factor-1 receptor antagonists for modulation of weight and liposity
WO2008076278A2 (en) 2006-12-13 2008-06-26 Schering Corporation Methods of cancer treatment with igf1r inhibitors
WO2008098917A2 (en) * 2007-02-14 2008-08-21 Glaxo Group Limited Novel antibodies against igf-ir
WO2008098917A3 (en) * 2007-02-14 2008-10-09 Glaxo Group Ltd Novel antibodies against igf-ir
JP2010518140A (en) * 2007-02-14 2010-05-27 グラクソ グループ リミテッド Novel antibody against IGF-1R
WO2008116103A3 (en) * 2007-03-22 2009-01-08 Imclone Systems Inc Stable antibody formulations
JP2010522208A (en) * 2007-03-22 2010-07-01 イムクローン・リミテッド・ライアビリティ・カンパニー Stable antibody formulation
US8492328B2 (en) 2007-05-17 2013-07-23 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
WO2008144345A2 (en) 2007-05-17 2008-11-27 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
EP2559771A2 (en) 2007-05-17 2013-02-20 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
US8178091B2 (en) 2007-05-21 2012-05-15 University Of Washington Compositions and methods for the treatment of respiratory disorders
WO2008152422A2 (en) * 2007-06-12 2008-12-18 Astrazeneca Ab Binding proteins specific for insulin-like growth factors and uses thereof-909
WO2008152422A3 (en) * 2007-06-12 2009-03-12 Astrazeneca Ab Binding proteins specific for insulin-like growth factors and uses thereof-909
US8580254B2 (en) 2007-06-19 2013-11-12 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
WO2009032782A2 (en) * 2007-08-28 2009-03-12 Biogen Idec Ma Inc. Compositions that bind multiple epitopes of igf-1r
WO2009032782A3 (en) * 2007-08-28 2009-05-28 Biogen Idec Inc Compositions that bind multiple epitopes of igf-1r
US8383114B2 (en) 2007-09-27 2013-02-26 Amgen Inc. Pharmaceutical formulations
US10653781B2 (en) 2007-09-27 2020-05-19 Amgen Inc. Pharmaceutical formulations
US9320797B2 (en) 2007-09-27 2016-04-26 Amgen Inc. Pharmaceutical formulations
EP3381445A2 (en) 2007-11-15 2018-10-03 Amgen Inc. Aqueous formulation of antibody stablised by antioxidants for parenteral administration
US8133489B2 (en) 2007-11-21 2012-03-13 Imclone Llc Inhibition of macrophage-stimulating protein receptor (RON) and methods of treatment thereof
US7947811B2 (en) 2007-11-21 2011-05-24 Imclone Llc Antibodies that bind specifically to human RON protein
WO2009080278A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Stability testing of antibodies
US9920108B2 (en) 2008-02-14 2018-03-20 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US10781247B2 (en) 2008-02-14 2020-09-22 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US9234028B2 (en) 2008-02-14 2016-01-12 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2009137378A3 (en) * 2008-05-05 2010-03-04 Schering Corporation Sequential administration of chemotherapeutic agents for treatment of cancer
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
US8318159B2 (en) 2008-12-12 2012-11-27 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US10179810B2 (en) 2008-12-12 2019-01-15 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US11299538B2 (en) 2008-12-12 2022-04-12 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
EP2236139A1 (en) 2009-03-31 2010-10-06 F. Hoffmann-La Roche AG Combination therapy of erlotinib with an anti-IGF-1R antibody, which does not inhibit binding of insulin to the insulin receptor
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2011050333A1 (en) 2009-10-23 2011-04-28 Amgen Inc. Vial adapter and system
US11136610B2 (en) 2009-10-26 2021-10-05 Hoffmann-La Roche Inc. Method for the production of a glycosylated immunoglobulin
US11021728B2 (en) 2009-10-26 2021-06-01 Hoffmann-La Roche Inc. Method for the production of a glycosylated immunoglobulin
US11377678B2 (en) 2009-10-26 2022-07-05 Hoffman-La Roche Inc. Method for the production of a glycosylated immunoglobulin
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2011053779A2 (en) 2009-10-30 2011-05-05 Bristol-Myers Squibb Company Methods for treating cancer in patients having igf-1r inhibitor resistance
WO2011083391A2 (en) 2010-01-05 2011-07-14 Pfizer Inc. Biomarkers for anti-igf-ir cancer therapy
WO2011101328A2 (en) 2010-02-18 2011-08-25 Roche Glycart Ag Treatment with a humanized igg class anti egfr antibody and an antibody against insulin like growth factor 1 receptor
US11161893B2 (en) 2010-05-26 2021-11-02 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US10273286B2 (en) 2010-05-26 2019-04-30 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US9562089B2 (en) 2010-05-26 2017-02-07 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
WO2011156373A1 (en) 2010-06-07 2011-12-15 Amgen Inc. Drug delivery device
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012106556A2 (en) 2011-02-02 2012-08-09 Amgen Inc. Methods and compositons relating to inhibition of igf-1r
WO2012135315A1 (en) 2011-03-31 2012-10-04 Amgen Inc. Vial adapter and system
EP3214096A3 (en) * 2011-04-19 2017-10-18 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-igf-1r and anti-erbb3 antibodies
US9938346B2 (en) 2011-04-19 2018-04-10 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-IGF-1R and anti-ErbB3 antibodies
CN105884900A (en) * 2011-04-19 2016-08-24 梅里麦克制药股份有限公司 Monospecific and bispecific anti-IGF-1R and anti-ERBB3 antibodies
EP4074355A1 (en) 2011-04-20 2022-10-19 Amgen Inc. Autoinjector apparatus
EP3498323A2 (en) 2011-04-20 2019-06-19 Amgen Inc. Autoinjector apparatus
WO2013056069A1 (en) 2011-10-13 2013-04-18 Bristol-Myers Squibb Company Methods for selecting and treating cancer in patients with igf-1r/ir inhibitors
EP3045187A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3045189A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3045190A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3045188A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3269413A1 (en) 2011-10-14 2018-01-17 Amgen, Inc Injector and method of assembly
EP3744371A1 (en) 2011-10-14 2020-12-02 Amgen, Inc Injector and method of assembly
EP3335747A1 (en) 2011-10-14 2018-06-20 Amgen Inc. Injector and method of assembly
WO2013055873A1 (en) 2011-10-14 2013-04-18 Amgen Inc. Injector and method of assembly
WO2013071056A2 (en) 2011-11-11 2013-05-16 Duke University Combination drug therapy for the treatment of solid tumors
US9700619B2 (en) 2011-11-11 2017-07-11 Duke University Combination drug therapy for the treatment of solid tumors
US9931402B2 (en) 2011-11-11 2018-04-03 Duke University Compositions for the treatment of solid tumors
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
EP2631653A1 (en) 2012-02-24 2013-08-28 Charité - Universitätsmedizin Berlin Identification of modulators of binding properties of antibodies reactive with a member of the insulin receptor family
WO2013124482A2 (en) 2012-02-24 2013-08-29 Charité - Universitätsmedizin Berlin Identification of modulators of binding properties of antibodies reactive with a member of the insulin receptor family
EP3527988A2 (en) 2012-02-24 2019-08-21 BerYsol GmbH Identification of antibodies reactive with a member of the insulin receptor family
WO2014015280A1 (en) 2012-07-20 2014-01-23 Novartis Pharma Ag Combination therapy of inhibitors for igf1 r and pi3k
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9163093B2 (en) 2012-11-01 2015-10-20 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
WO2014067642A1 (en) 2012-11-05 2014-05-08 Mab Discovery Gmbh Method for the production of multispecific antibodies
EP2727941A1 (en) 2012-11-05 2014-05-07 MAB Discovery GmbH Method for the production of multispecific antibodies
EP3081249A1 (en) 2012-11-21 2016-10-19 Amgen, Inc Drug delivery device
EP3656426A1 (en) 2012-11-21 2020-05-27 Amgen, Inc Drug delivery device
US10682474B2 (en) 2012-11-21 2020-06-16 Amgen Inc. Drug delivery device
EP3072548A1 (en) 2012-11-21 2016-09-28 Amgen, Inc Drug delivery device
US11439745B2 (en) 2012-11-21 2022-09-13 Amgen Inc. Drug delivery device
US11344681B2 (en) 2012-11-21 2022-05-31 Amgen Inc. Drug delivery device
EP4234694A2 (en) 2012-11-21 2023-08-30 Amgen Inc. Drug delivery device
WO2014081780A1 (en) 2012-11-21 2014-05-30 Amgen Inc. Drug delivery device
US11458247B2 (en) 2012-11-21 2022-10-04 Amgen Inc. Drug delivery device
US10377828B2 (en) 2013-03-07 2019-08-13 Boehringer Ingelheim International Gmbh Combination therapy for neoplasia treatment
US9062108B2 (en) 2013-03-15 2015-06-23 Abbvie Inc. Dual specific binding proteins directed against IL-1 and/or IL-17
WO2014144096A1 (en) 2013-03-15 2014-09-18 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
WO2014149357A1 (en) 2013-03-22 2014-09-25 Amgen Inc. Injector and method of assembly
EP3831427A1 (en) 2013-03-22 2021-06-09 Amgen Inc. Injector and method of assembly
EP3501575A1 (en) 2013-10-24 2019-06-26 Amgen, Inc Drug delivery system with temperature-sensitive-control
EP3421066A1 (en) 2013-10-24 2019-01-02 Amgen, Inc Injector and method of assembly
WO2015061389A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Drug delivery system with temperature-sensitive control
WO2015061386A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Injector and method of assembly
EP3789064A1 (en) 2013-10-24 2021-03-10 Amgen, Inc Injector and method of assembly
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
EP3785749A1 (en) 2014-05-07 2021-03-03 Amgen Inc. Autoinjector with shock reducing elements
WO2015171777A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
US11213624B2 (en) 2014-06-03 2022-01-04 Amgen Inc. Controllable drug delivery system and method of use
EP4036924A1 (en) 2014-06-03 2022-08-03 Amgen, Inc Devices and methods for assisting a user of a drug delivery device
WO2015187797A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Controllable drug delivery system and method of use
WO2015187799A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Systems and methods for remotely processing data collected by a drug delivery device
WO2015187793A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Drug delivery system and method of use
US11738146B2 (en) 2014-06-03 2023-08-29 Amgen Inc. Drug delivery system and method of use
EP3943135A2 (en) 2014-10-14 2022-01-26 Amgen Inc. Drug injection device with visual and audible indicators
WO2016061220A2 (en) 2014-10-14 2016-04-21 Amgen Inc. Drug injection device with visual and audio indicators
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
US11357916B2 (en) 2014-12-19 2022-06-14 Amgen Inc. Drug delivery device with live button or user interface field
US11944794B2 (en) 2014-12-19 2024-04-02 Amgen Inc. Drug delivery device with proximity sensor
US10765801B2 (en) 2014-12-19 2020-09-08 Amgen Inc. Drug delivery device with proximity sensor
EP3848072A1 (en) 2014-12-19 2021-07-14 Amgen Inc. Drug delivery device with proximity sensor
WO2016100055A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with live button or user interface field
WO2016100781A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with proximity sensor
EP3689394A1 (en) 2014-12-19 2020-08-05 Amgen Inc. Drug delivery device with live button or user interface field
US10799630B2 (en) 2014-12-19 2020-10-13 Amgen Inc. Drug delivery device with proximity sensor
WO2016133947A1 (en) 2015-02-17 2016-08-25 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
EP3556411A1 (en) 2015-02-17 2019-10-23 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
EP3981450A1 (en) 2015-02-27 2022-04-13 Amgen, Inc Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
WO2016138434A1 (en) 2015-02-27 2016-09-01 Amgen Inc. Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
WO2017100501A1 (en) 2015-12-09 2017-06-15 Amgen Inc. Auto-injector with signaling cap
WO2017120178A1 (en) 2016-01-06 2017-07-13 Amgen Inc. Auto-injector with signaling electronics
EP4035711A1 (en) 2016-03-15 2022-08-03 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
WO2017160799A1 (en) 2016-03-15 2017-09-21 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
EP3721922A1 (en) 2016-03-15 2020-10-14 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
WO2017189089A1 (en) 2016-04-29 2017-11-02 Amgen Inc. Drug delivery device with messaging label
WO2017192287A1 (en) 2016-05-02 2017-11-09 Amgen Inc. Syringe adapter and guide for filling an on-body injector
WO2017197222A1 (en) 2016-05-13 2017-11-16 Amgen Inc. Vial sleeve assembly
WO2017200989A1 (en) 2016-05-16 2017-11-23 Amgen Inc. Data encryption in medical devices with limited computational capability
WO2017209899A1 (en) 2016-06-03 2017-12-07 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
WO2018004842A1 (en) 2016-07-01 2018-01-04 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
WO2018081234A1 (en) 2016-10-25 2018-05-03 Amgen Inc. On-body injector
WO2018136398A1 (en) 2017-01-17 2018-07-26 Amgen Inc. Injection devices and related methods of use and assembly
WO2018151890A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Drug delivery device with sterile fluid flowpath and related method of assembly
WO2018152073A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Insertion mechanism for drug delivery device
WO2018165143A1 (en) 2017-03-06 2018-09-13 Amgen Inc. Drug delivery device with activation prevention feature
WO2018164829A1 (en) 2017-03-07 2018-09-13 Amgen Inc. Needle insertion by overpressure
WO2018165499A1 (en) 2017-03-09 2018-09-13 Amgen Inc. Insertion mechanism for drug delivery device
WO2018183039A1 (en) 2017-03-28 2018-10-04 Amgen Inc. Plunger rod and syringe assembly system and method
EP4241807A2 (en) 2017-03-28 2023-09-13 Amgen Inc. Plunger rod and syringe assembly system and method
WO2018226515A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
WO2018226565A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Torque driven drug delivery device
WO2018236619A1 (en) 2017-06-22 2018-12-27 Amgen Inc. Device activation impact/shock reduction
WO2018237225A1 (en) 2017-06-23 2018-12-27 Amgen Inc. Electronic drug delivery device comprising a cap activated by a switch assembly
WO2019014014A1 (en) 2017-07-14 2019-01-17 Amgen Inc. Needle insertion-retraction system having dual torsion spring system
WO2019018169A1 (en) 2017-07-21 2019-01-24 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
EP4292576A2 (en) 2017-07-21 2023-12-20 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
EP4085942A1 (en) 2017-07-25 2022-11-09 Amgen Inc. Drug delivery device with gear module and related method of assembly
WO2019022950A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with container access system and related method of assembly
WO2019022951A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with gear module and related method of assembly
WO2019032482A2 (en) 2017-08-09 2019-02-14 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
WO2019036181A1 (en) 2017-08-18 2019-02-21 Amgen Inc. Wearable injector with sterile adhesive patch
WO2019040548A1 (en) 2017-08-22 2019-02-28 Amgen Inc. Needle insertion mechanism for drug delivery device
WO2019070472A1 (en) 2017-10-04 2019-04-11 Amgen Inc. Flow adapter for drug delivery device
WO2019070552A1 (en) 2017-10-06 2019-04-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
EP4257164A2 (en) 2017-10-06 2023-10-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
WO2019074579A1 (en) 2017-10-09 2019-04-18 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
WO2019090086A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Systems and approaches for sterilizing a drug delivery device
WO2019090079A1 (en) 2017-11-03 2019-05-09 Amgen Inc. System and approaches for sterilizing a drug delivery device
WO2019089178A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Drug delivery device with placement and flow sensing
WO2019090303A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Fill-finish assemblies and related methods
WO2019094138A1 (en) 2017-11-10 2019-05-16 Amgen Inc. Plungers for drug delivery devices
WO2019099324A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Door latch mechanism for drug delivery device
WO2019099322A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Autoinjector with stall and end point detection
WO2019231582A1 (en) 2018-05-30 2019-12-05 Amgen Inc. Thermal spring release mechanism for a drug delivery device
WO2019231618A1 (en) 2018-06-01 2019-12-05 Amgen Inc. Modular fluid path assemblies for drug delivery devices
WO2020023451A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
WO2020023444A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020028009A1 (en) 2018-07-31 2020-02-06 Amgen Inc. Fluid path assembly for a drug delivery device
WO2020068623A1 (en) 2018-09-24 2020-04-02 Amgen Inc. Interventional dosing systems and methods
WO2020068476A1 (en) 2018-09-28 2020-04-02 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
WO2020072577A1 (en) 2018-10-02 2020-04-09 Amgen Inc. Injection systems for drug delivery with internal force transmission
WO2020072846A1 (en) 2018-10-05 2020-04-09 Amgen Inc. Drug delivery device having dose indicator
WO2020081479A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Drug delivery device having damping mechanism
WO2020081480A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Platform assembly process for drug delivery device
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020092056A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial needle retraction
WO2020091956A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020219482A1 (en) 2019-04-24 2020-10-29 Amgen Inc. Syringe sterilization verification assemblies and methods
WO2021041067A2 (en) 2019-08-23 2021-03-04 Amgen Inc. Drug delivery device with configurable needle shield engagement components and related methods
WO2022246055A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container
WO2023138620A1 (en) * 2022-01-19 2023-07-27 黄浩旻 Anti-pdl1/igf1r bispecific antibody, preparation method therefor and use thereof

Also Published As

Publication number Publication date
WO2005016970A3 (en) 2005-04-21
US20080025990A1 (en) 2008-01-31
EP1622942A2 (en) 2006-02-08
JP2011184440A (en) 2011-09-22
US20100158920A1 (en) 2010-06-24
JP2007535895A (en) 2007-12-13
CA2524305C (en) 2015-12-08
EP2365001A3 (en) 2012-03-28
EP1622942B1 (en) 2014-11-19
ES2527871T3 (en) 2015-02-02
CA2524305A1 (en) 2005-02-24
EP2365001A2 (en) 2011-09-14
JP5638988B2 (en) 2014-12-10
US7968093B2 (en) 2011-06-28
US7638605B2 (en) 2009-12-29

Similar Documents

Publication Publication Date Title
US7968093B2 (en) Fully human antibodies directed against the human insulin-like growth factor-1 receptor
CA2616099C (en) Novel anti-igf-ir antibodies and uses thereof
KR101246428B1 (en) Methods of Treating a Secondary Bone Tumor with Anti-PDGFRalpha Antibodies
CA2560305C (en) Human anti-epidermal growth factor receptor antibody
JP5723279B2 (en) Anti-CXCR4 antibodies and their use for cancer treatment
US8071730B2 (en) Anti-JAM-A antibodies
EP2300500A1 (en) Anti-flt3 antibodies

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2524305

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006514278

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2004775941

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/001458

Country of ref document: MX

WWP Wipo information: published in national office

Ref document number: 2004775941

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10555407

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10555407

Country of ref document: US